EP4156909A1 - Procédés de régulation de la taille du méristème pour l'amélioration des cultures - Google Patents

Procédés de régulation de la taille du méristème pour l'amélioration des cultures

Info

Publication number
EP4156909A1
EP4156909A1 EP21735063.6A EP21735063A EP4156909A1 EP 4156909 A1 EP4156909 A1 EP 4156909A1 EP 21735063 A EP21735063 A EP 21735063A EP 4156909 A1 EP4156909 A1 EP 4156909A1
Authority
EP
European Patent Office
Prior art keywords
sequence
plant
crn
gene
seq
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
EP21735063.6A
Other languages
German (de)
English (en)
Inventor
Devin O'CONNOR
Nathaniel GRAHAM
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Pairwise Plants Services Inc
Original Assignee
Pairwise Plants Services Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Pairwise Plants Services Inc filed Critical Pairwise Plants Services Inc
Publication of EP4156909A1 publication Critical patent/EP4156909A1/fr
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01HNEW PLANTS OR NON-TRANSGENIC PROCESSES FOR OBTAINING THEM; PLANT REPRODUCTION BY TISSUE CULTURE TECHNIQUES
    • A01H6/00Angiosperms, i.e. flowering plants, characterised by their botanic taxonomy
    • A01H6/46Gramineae or Poaceae, e.g. ryegrass, rice, wheat or maize
    • A01H6/4684Zea mays [maize]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/82Vectors or expression systems specially adapted for eukaryotic hosts for plant cells, e.g. plant artificial chromosomes (PACs)
    • C12N15/8241Phenotypically and genetically modified plants via recombinant DNA technology
    • C12N15/8261Phenotypically and genetically modified plants via recombinant DNA technology with agronomic (input) traits, e.g. crop yield
    • C12N15/8262Phenotypically and genetically modified plants via recombinant DNA technology with agronomic (input) traits, e.g. crop yield involving plant development
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01HNEW PLANTS OR NON-TRANSGENIC PROCESSES FOR OBTAINING THEM; PLANT REPRODUCTION BY TISSUE CULTURE TECHNIQUES
    • A01H1/00Processes for modifying genotypes ; Plants characterised by associated natural traits
    • A01H1/12Processes for modifying agronomic input traits, e.g. crop yield
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/82Vectors or expression systems specially adapted for eukaryotic hosts for plant cells, e.g. plant artificial chromosomes (PACs)
    • C12N15/8201Methods for introducing genetic material into plant cells, e.g. DNA, RNA, stable or transient incorporation, tissue culture methods adapted for transformation
    • C12N15/8213Targeted insertion of genes into the plant genome by homologous recombination
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/82Vectors or expression systems specially adapted for eukaryotic hosts for plant cells, e.g. plant artificial chromosomes (PACs)
    • C12N15/8241Phenotypically and genetically modified plants via recombinant DNA technology
    • C12N15/8261Phenotypically and genetically modified plants via recombinant DNA technology with agronomic (input) traits, e.g. crop yield
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/16Hydrolases (3) acting on ester bonds (3.1)
    • C12N9/22Ribonucleases RNAses, DNAses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/20Type of nucleic acid involving clustered regularly interspaced short palindromic repeats [CRISPRs]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2800/00Nucleic acids vectors
    • C12N2800/80Vectors containing sites for inducing double-stranded breaks, e.g. meganuclease restriction sites
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A40/00Adaptation technologies in agriculture, forestry, livestock or agroalimentary production
    • Y02A40/10Adaptation technologies in agriculture, forestry, livestock or agroalimentary production in agriculture
    • Y02A40/146Genetically Modified [GMO] plants, e.g. transgenic plants

Definitions

  • This invention relates to compositions and methods for modifying CORYNE (CRN) genes in plants, optionally to increase kernel row number.
  • the invention further relates to plants having a modified CRN gene and, for example, increased kernel row number produced using the methods and compositions of the invention.
  • New plant organs are initiated at the growing tip of the plant called the meristem.
  • the meristem a population of undifferentiated stem cells is maintained.
  • the meristem allocates stem-cells to newly formed organs, including seeds, while at the same time reserving some stem-cells to continually maintain the meristem.
  • kernel row number As a result of the modular nature of maize ear development, larger meristems tend to initiate more flowers, and thus, meristem size has a direct effect on kernel row number and yield.
  • the number of flowers initiated during the development of the maize ear directly limits grain yield.
  • An increased number of flowers initiated around the circumference of the ear was a major trait selected during maize domestication.
  • Significant advancements through breeding have resulted in dramatic increases in kernel row number, from 2 in teosinte, the ancestor of maize, to -8-20 rows in modem elite maize varieties. In diverse maize lines kernel row number can get as high as 36.
  • CLAVATA3 CLV3 peptide is secreted from cells in the meristem apex and moves through the apoplast into the central stem-cell domain where it interacts with several Leucine Rich Receptors (LRRs) including CLAVATA1 (CLV1) and CLAVATA2 (CLV2).
  • LRRs Leucine Rich Receptors
  • CLV1 CLV1
  • CLAVATA2 CLV2
  • This receptor- ligand interaction stimulates signaling that ultimately acts to reduce WUS expression and restrict the expansion of the stem cell population.
  • One of the targets of WUS is the CLV3 gene itself, and in this way WUS acts to limit its own expression and maintain stem cell homeostasis (Fletcher, J.C Plants 7: 87 (2016)).
  • CLV-WUS signaling is transduced downstream via CRN. While strong loss-of-function mutations in the maize CLV2 ortholog FACIATED EAR2 (FEA2) result in enlarged meristems and an increase in KRN, the ear is disordered and as a result there is no yield increase (Taguchi-Shiobara et al., Gene Dev 15:2755-2766 (2001)).
  • One aspect of the invention provides a plant or plant part thereof comprising at least one non-natural mutation in a short extracellular (EC) domain of an endogenous CORYNE (CRN) gene that encodes a CRN protein.
  • EC extracellular
  • CRN CORYNE
  • a second aspect of the invention provides a plant cell, comprising an editing system comprising: (a) a CRISPR-Cas effector protein; and (b) a guide nucleic acid (gRNA, gDNA, crRNA, crDNA, sgRNA, sgDNA) comprising a spacer sequence with complementarity to an endogenous target gene encoding an CRN protein.
  • an editing system comprising: (a) a CRISPR-Cas effector protein; and (b) a guide nucleic acid (gRNA, gDNA, crRNA, crDNA, sgRNA, sgDNA) comprising a spacer sequence with complementarity to an endogenous target gene encoding an CRN protein.
  • a third aspect of the invention provides a com plant cell comprising at least one non- naturally occurring mutation within a CRN gene, wherein the mutation is a substitution, insertion or a deletion that is introduced using an editing system that comprises a nucleic acid binding domain that binds to a target site in the CRN gene.
  • a fourth aspect of the invention provides a method of producing/breeding a transgene- free edited com plant, comprising: crossing the com plant of the invention with a transgene free com plant, thereby introducing the at least one non-natural mutation into the com plant that is transgene-free; and selecting a progeny com plant that comprises the at least one non-natural mutation and is transgene-free, thereby producing a transgene free edited com plant.
  • a fifth aspect of the invention provides a method of providing a plurality of com plants having increased kernel number, the method comprising planting two or more plants of the invention in a growing area, thereby providing a plurality of com plants having increased kernel number as compared to a plurality of control com plants not comprising the mutation.
  • a sixth aspect of the invention provides a method of generating variation in a region of a com CRN protein, comprising: introducing an editing system into a com plant cell, wherein the editing system is targeted to a region of a com CRN gene that encodes the region of the com CRN protein, wherein the region comprises a sequence having at least 70% sequence identity (e.g., 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84 ,85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100%) to any one of the amino acid sequences of SEQ ID NO: 119-121 or the region is encoded by a sequence having at least 70% identity to the nucleotide sequence of SEQ ID NOs: 123-125; and contacting the region of the CRN gene with the editing system, generating variation in
  • a seventh aspect of the invention provides a method for editing a specific site in the genome of a plant cell, the method comprising: cleaving, in a site specific manner, a target site within an endogenous CRN gene in the plant cell, the endogenous CRN gene comprising a sequence having at least 70% sequence identity to the nucleotide sequence of SEQ ID NO:122, or encoding a sequence having at least 70% sequence identity to the amino acid sequence of SEQ ID NO:118, thereby generating an edit in the endogenous CRN gene of the plant cell and producing a plant cell comprising the edit in the endogenous CRN gene.
  • An eighth aspect provides a method for making a com plant, comprising: (a) contacting a population of com plant cells comprising a wild-type endogenous CRN gene with a nuclease linked to a nucleic acid binding domain (e.g., DNA binding domain, e.g., editing system) that binds to a sequence having at least 70% sequence identity to the nucleotide sequence of SEQ ID NO:122, to a sequence having at least 70% identity to the nucleotide sequence of SEQ ID
  • a ninth aspect provides a method for increasing kernel number in a com plant, comprising (a) contacting a com plant cell comprising an endogenous CRN gene with a nuclease targeting the endogenous CRN gene, wherein the nuclease is linked to a nucleic acid binding domain (e.g., DNA binding domain, e.g., editing system) that binds to a target site in the endogenous CRN gene, wherein the endogenous CRN gene: (i) encodes a sequence having at least 70% sequence identity to the amino acid sequence of SEQ ID NO:118; (ii) comprises a sequence having at least 70% sequence identity to the nucleotide sequence of SEQ ID NO:122; (iii) comprises a region having a sequence with at least 70% sequence identity to the nucleotide sequence of SEQ ID NOs:123-125, optionally SEQ ID NO:125; and/or (iv) comprises a region encoding a sequence having at least 70% sequence identity to any one of
  • a tenth aspect provides method for producing a com plant or part thereof comprising at least one cell having a mutated endogenous CRN gene, the method comprising contacting a target site in an endogenous CRN gene in the com plant or plant part with a nuclease comprising a cleavage domain and a nucleic acid binding domain, wherein the nucleic acid binding domain binds to a target site in the endogenous CRN gene, wherein the endogenous CRN gene (a) encodes a sequence having at least 70% sequence identity to the amino acid sequence of SEQ ID NO:118; (b) comprises a sequence having at least 70% sequence identity to the nucleotide sequence of SEQ ID NO:122; (c) comprises a region having a sequence with at least 70% sequence identity to the nucleotide sequence of SEQ ID NOs: 123-125, optionally SEQ ID NO: 125; and/or (d) comprises a region encoding a sequence having at least 70% sequence identity to the amino acid sequence
  • An eleventh aspect of the invention provides a method for producing a com plant or part thereof comprising a mutated endogenous CRN gene and exhibiting increased kernel number, the method comprising contacting a target site in an endogenous CRN gene in the com plant or plant part with a nuclease comprising a cleavage domain and a nucleic acid binding domain, wherein the nucleic acid binding domain binds to a target site in the endogenous CRN gene, wherein the endogenous CRN gene: (a) encodes a sequence having at least 70% sequence identity to the amino acid sequence of SEQ ID NO:118; (b) comprises a sequence having at least 70% sequence identity to the nucleotide sequence of SEQ ID NO:122; (c) comprises a region having a sequence with at least 70% sequence identity to the nucleotide sequence of SEQ ID NOs: 123-125, optionally SEQ ID NO: 125; and/or (d) comprises a region encoding a sequence having at least 70% sequence
  • a twelfth aspect provides a guide nucleic acid that binds to a target site in a CRN gene, the target site comprising a sequence having at least 70% sequence identity to the nucleotide sequence of SEQ ID NO:122, at least 70% sequence identity to the nucleotide sequence of SEQ ID NO:123-125, optionally SEQ ID NO:125; encoding a sequence having at least 70% sequence identity to the amino acid sequence of SEQ ID NO:118, or encoding a sequence having at least 70% sequence identity to any one of the amino acid sequences of SEQ ID NO:119-121.
  • a system comprising a guide nucleic acid of the invention and a CRISPR-Cas effector protein that associates with the guide nucleic acid.
  • a fourteenth aspect provides a gene editing system comprising a CRISPR-Cas effector protein in association with a guide nucleic acid, wherein the guide nucleic acid comprises a spacer sequence that binds to an endogenous CRN gene.
  • a complex comprising a CRISPR-Cas effector protein comprising a cleavage domain and a guide nucleic acid, wherein the guide nucleic acid binds to a target site in an endogenous CRN gene, wherein the endogenous CRN gene, wherein the endogenous CRN gene, wherein the endogenous CRN gene: (a) encodes a sequence having at least 70% sequence identity to the amino acid sequence of SEQ ID NO:118; (b) comprises a sequence having at least 70% sequence identity to the nucleotide sequence of SEQ ID NO:122; (c) comprises a region having a sequence with at least 70% sequence identity to the nucleotide sequence of any one of SEQ ID NO:123-125, optionally SEQ ID NO: 125; and/or (d) comprises a region encoding a sequence having at least 70% sequence identity to any one of the amino acid sequences of SEQ ID NOs:119-121, wherein the cleavage domain cleaves a target strand in the
  • an expression cassette comprising (a) a polynucleotide encoding CRISPR-Cas effector protein comprising a cleavage domain and (b) a guide nucleic acid that binds to a target site in an endogenous CRN gene, wherein the guide nucleic acid comprises a spacer sequence that is complementary to and binds to (i) a portion of a nucleic acid encoding an amino acid sequence having at least 70% sequence identity the amino acid sequence of SEQ ID NO: 118; (ii) a portion of a sequence having at least 70% sequence identity to the nucleotide sequence of SEQ ID NO:122; (iii) a portion of a sequence having at least 70% sequence identity to any one of the nucleotide sequences of SEQ ID NO:123-125, optionally SEQ ID NO:125; and/or (iv) a portion of sequence having at least 70% sequence identity to a sequence encoding any one of the amino acid sequence
  • a method of creating a mutation in an endogenous CRN gene in a plant comprising: (a) targeting a gene editing system to a portion of the CRN gene, the portion comprising (i) a sequence having at least 70% sequence identity to any one of the nucleotide sequences of SEQ ID NOs:123-125, optionally SEQ ID NO:125; and/or (ii) a sequence having at least 70% sequence identity to a sequence encoding any one of the amino acid sequences of SEQ ID NO: 118-121, and (b) selecting a plant that comprises a substitution of an amino acid residue in the EC domain of the CRN gene, optionally an alternative amino acid in amino acid residue at position 477.
  • a further aspect of the invention provides a nucleic acid encoding a dominant negative mutation, a semi-dominant mutation, a hypomorphic mutation, or a weak loss-of-function mutation of a com CRN protein.
  • a com plant or part thereof comprising a nucleic acid of the invention.
  • a com plant or part thereof is provided that exhibits increased kernel number.
  • a com plant is provided that also exhibits increased yield, and improved disease resistance as well as exhibits larger meristems and root meristems that are maintained.
  • plants comprising in their genome one or more mutated CORYNE (CRN) genes produced by the methods of the invention as well as polypeptides, polynucleotides, nucleic acid constmcts, expression cassettes and vectors for making a plant of this invention.
  • CRN mutated CORYNE
  • SEQ ID NOs:l-17 are exemplary Casl2a amino acid sequences useful with this invention.
  • SEQ ID NOs:18-20 are exemplary Cas 12a nucleotide sequences useful with this invention.
  • SEQ ID NO:21-22 are exemplary regulatory sequences encoding a promoter and intron.
  • SEQ ID NOs:23-29 are exemplary cytosine deaminase sequences useful with this invention.
  • SEQ ID N0s:30-40 are exemplary adenine deaminase amino acid sequences useful with this invention.
  • SEQ ID NO:41 is an exemplary uracil-DNA glycosylase inhibitor (UGI) sequences useful with this invention.
  • SEQ ID NOs:42-44 provides an example of a protospacer adjacent motif position for a Type V CRISPR-Cas 12a nuclease.
  • SEQ ID NOs:45-47 provide example peptide tags and affinity polypeptides useful with this invention.
  • SEQ ID NOs:48-58 provide example RNA recruiting motifs and corresponding affinity polypeptides useful with this invention.
  • SEQ ID NOs:59-60 are exemplary Cas9 polypeptide sequences useful with this invention.
  • SEQ ID NOs:61-71 are exemplary Cas9 polynucleotide sequences useful with this invention.
  • SEQ ID Nos:72-118 are example CRN polypeptide sequences.
  • SEQ ID NO: 119 is an example EC domain amino acid sequence from a maize CRN polypeptide.
  • SEQ ID NO: 120 and SEQ ID NO: 121 are example target regions of a CRN polypeptide.
  • SEQ ID NO: 122 is an example CRN genomic sequence.
  • SEQ ID NOs: 123-125 are example target regions in the CRN genomic sequence.
  • SEQ ID NOs: 126-130 are example spacer sequences for nucleic acid guides useful with this invention.
  • SEQ ID Nos: 131-178 are the sequences shown in Fig. 1 from top to bottom.
  • SEQ ID NO: 179 is a portion of the wildtype CRN sequence as shown in Fig. 2, top line.
  • SEQ ID NOs: 180-191 show example edits of a CRN nucleotide sequence as shown in Fig. 2, second line to bottom line.
  • FIG. 1 provides an alignment of various CRN polypeptides showing the EC domain (from top to bottom: SEQ ID NOs: 131-178).
  • FIG. 2 provides an alignment of mutated sequences with the wild type CRN sequence (from top to bottom: SEQ ID NO: 179-191).
  • FIG. 3 provides a cartoon exemplifying a structure of wild type (WT) maize CRN protein and edited maize CRN proteins showing in-frame deletions.
  • FIG. 4 provides a cartoon exemplifying a maize CRN polypeptide functioning in planta.
  • a measurable value such as an amount or concentration and the like, is meant to encompass variations of ⁇ 10%, ⁇ 5%, ⁇ 1%, ⁇ 0.5%, or even ⁇ 0.1% of the specified value as well as the specified value.
  • "about X" where X is the measurable value is meant to include X as well as variations of ⁇ 10%, ⁇ 5%, ⁇ 1%, ⁇ 0.5%, or even ⁇ 0.1% of X.
  • a range provided herein for a measurable value may include any other range and/or individual value therein.
  • phrases such as “between X and Y” and “between about X and Y” should be interpreted to include X and Y.
  • phrases such as “between about X and Y” mean “between about X and about Y” and phrases such as “from about X to Y” mean “from about X to about Y.”
  • the terms “increase,” “increasing,” “increased,” “enhance,” “enhanced,” “enhancing,” and “enhancement” (and grammatical variations thereof) describe an elevation of at least about 5%, 10%, 15%, 20%, 25%, 50%, 75%, 100%, 150%, 200%, 300%, 400%, 500% or more as compared to a control.
  • a plant comprising a mutation in a CRN gene as described herein can exhibit increased kernel row number (e.g., producing ears having increased kernel row number) that is at least about 5% or greater than that of a control plant not comprising the same mutation, optionally wherein the length of the ears comprising increased kernel row number is not substantially decreased (e.g., a decrease in length of less than 30% as compared to an ear of a plant not comprising the same FEA2 mutation).
  • a control plant is typically the same plant as the edited plant but the control plant has not been similarly edited and therefore does not comprise the mutation.
  • a control plant maybe an isogenic plant and/or a wild type plant.
  • a control plant can be the same breeding line, variety, or cultivar as the subject plant into which a mutation as described herein is introgressed, but the control breeding line, variety, or cultivar is free of the mutation.
  • a comparison between a plant of the invention and a control plant is made under the same growth conditions, e.g., the same environmental conditions (soil, hydration, light, heat, nutrients and the like).
  • a “control” plant maybe an isogenic plant and/or a wild type plant.
  • a control plant can be the same breeding line, variety, or cultivar as the subject plant into which a mutation as described herein is introgressed, but the control breeding line, variety, or cultivar is free of the mutation.
  • a comparison between a plant of the invention and a control plant is made under the same growth conditions, e.g., the same environmental conditions (soil, hydration, light, heat, nutrients and the like).
  • the terms “reduce,” “reduced,” “reducing,” “reduction,” “diminish,” and “decrease” describe, for example, a decrease of at least about 5%, 10%, 15%, 20%, 25%, 35%, 50%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%,
  • the reduction can result in no or essentially no (i.e., an insignificant amount, e.g., less than about 10% or even 5%) detectable activity or amount.
  • the terms "express,” “expresses,” “expressed” or “expression,” and the like, with respect to a nucleic acid molecule and/or a nucleotide sequence indicates that the nucleic acid molecule and/or a nucleotide sequence is transcribed and, optionally, translated.
  • a nucleic acid molecule and/or a nucleotide sequence may express a polypeptide of interest or, for example, a functional untranslated RNA.
  • a “heterologous” or a “recombinant” nucleotide sequence is a nucleotide sequence not naturally associated with a host cell into which it is introduced, including non- naturally occurring multiple copies of a naturally occurring nucleotide sequence.
  • a “heterologous” nucleotide/polypeptide may originate from a foreign species, or, if from the same species, is substantially modified from its native form in composition and/or genomic locus by deliberate human intervention.
  • a “native” or “wild type” nucleic acid, nucleotide sequence, polypeptide or amino acid sequence refers to a naturally occurring or endogenous nucleic acid, nucleotide sequence, polypeptide or amino acid sequence.
  • a "wild type” nucleic acid is a nucleic acid that is not edited as described herein and can differ from an "endogenous" gene that may be edited as described herein (e.g., a mutated endogenous gene).
  • a "wild type” nucleic acid e.g., unedited
  • may be heterologous to the organism in which the wild type nucleic acid is found e.g., a transgenic organism).
  • a wild type endogenous CORYNE (CRN) gene is an CRN gene that is naturally occurring in or endogenous to the reference organism, e.g., a plant, e.g., a maize plant, and may be subject to modification as described herein, after which, such a modified endogenous gene is no longer wild type.
  • heterozygous refers to a genetic status wherein different alleles reside at corresponding loci on homologous chromosomes.
  • homozygous refers to a genetic status wherein identical alleles reside at corresponding loci on homologous chromosomes.
  • allele refers to one of two or more different nucleotides or nucleotide sequences that occur at a specific locus.
  • a "null allele” is a nonfunctional allele caused by a genetic mutation that results in a complete lack of production of the corresponding protein or produces a protein that is non functional.
  • a “dominant negative mutation” is a mutation that produces an altered gene product (e.g., having an aberrant function relative to wild type), which gene product adversely affects the function of the wild-type allele or gene product.
  • a “dominant negative mutation” may block a function of the wild type gene product.
  • a dominant negative mutation may also be referred to as an "antimorphic mutation.”
  • a “semi-dominant mutation” refers to a mutation in which the penetrance of the phenotype in a heterozygous organism is less than that observed for a homozygous organism.
  • a "weak loss-of-function mutation” is a mutation that results in a gene product having partial function or reduced function (partially inactivated) as compared to the wildtype gene product.
  • a “hypo orphic mutation” is a mutation that results in a partial loss of gene function which may occur through reduced expression (e.g., reduced protein and/or reduced RNA) or reduced functional performance (e.g , reduced activity), but not a complete loss of function/activity.
  • a “hypomorphic” allele is a semi-functional allele caused by a genetic mutation that results in production of the corresponding protein that functions at anywhere between 1% and 99% of normal efficiency.
  • a “hypermorphic mutation” is a mutation that results in increased expression of the gene product and/or increased activity of the gene product.
  • locus is a position on a chromosome where a gene or marker or allele is located. In some embodiments, a locus may encompass one or more nucleotides.
  • a desired allele As used herein, the terms “desired allele,” “target allele” and/or “allele of interest” are used interchangeably to refer to an allele associated with a desired trait.
  • a desired allele may be associated with either an increase or a decrease (relative to a control) of or in a given trait, depending on the nature of the desired phenotype.
  • a marker is "associated with” a trait when said trait is linked to it and when the presence of the marker is an indicator of whether and/or to what extent the desired trait or trait form will occur in a plant/germplasm comprising the marker.
  • a marker is "associated with” an allele or chromosome interval when it is linked to it and when the presence of the marker is an indicator of whether the allele or chromosome interval is present in a plant/germplasm comprising the marker.
  • backcross and “backcrossing” refer to the process whereby a progeny plant is crossed back to one of its parents one or more times (e.g., 1, 2, 3, 4, 5, 6, 7, 8, etc.).
  • the "donor” parent refers to the parental plant with the desired gene or locus to be introgressed.
  • the “recipient” parent (used one or more times) or “recurrent” parent (used two or more times) refers to the parental plant into which the gene or locus is being introgressed. For example, see Ragot, M. et al. Marker-assisted Backcrossing: A Practical
  • cross refers to the fusion of gametes via pollination to produce progeny (e.g., cells, seeds or plants).
  • progeny e.g., cells, seeds or plants.
  • the term encompasses both sexual crosses (the pollination of one plant by another) and selfing (self-pollination, e.g., when the pollen and ovule are from the same plant).
  • crossing refers to the act of fusing gametes via pollination to produce progeny.
  • a desired allele at a specified locus can be transmitted to at least one progeny via a sexual cross between two parents of the same species, where at least one of the parents has the desired allele in its genome.
  • transmission of an allele can occur by recombination between two donor genomes, e.g., in a fused protoplast, where at least one of the donor protoplasts has the desired allele in its genome.
  • the desired allele may be a selected allele of a marker, a QTL, a transgene, or the like.
  • Offspring comprising the desired allele can be backcrossed one or more times (e.g., 1, 2, 3, 4, or more times) to a line having a desired genetic background, selecting for the desired allele, with the result being that the desired allele becomes fixed in the desired genetic background.
  • a marker associated with increased yield under non-water stress conditions may be introgressed from a donor into a recurrent parent that does not comprise the marker and does not exhibit increased yield under non-water stress conditions.
  • the resulting offspring could then be backcrossed one or more times and selected until the progeny possess the genetic marker(s) associated with increased yield under non-water stress conditions in the recurrent parent background.
  • a "genetic map” is a description of genetic linkage relationships among loci on one or more chromosomes within a given species, generally depicted in a diagrammatic or tabular form. For each genetic map, distances between loci are measured by the recombination frequencies between them. Recombination between loci can be detected using a variety of markers.
  • a genetic map is a product of the mapping population, types of markers used, and the polymorphic potential of each marker between different populations. The order and genetic distances between loci can differ from one genetic map to another.
  • genotype refers to the genetic constitution of an individual (or group of individuals) at one or more genetic loci, as contrasted with the observable and/or detectable and/or manifested trait (the phenotype).
  • Genotype is defined by the allele(s) of one or more known loci that the individual has inherited from its parents.
  • genotype can be used to refer to an individual's genetic constitution at a single locus, at multiple loci, or more generally, the term genotype can be used to refer to an individual's genetic make-up for all the genes in its genome. Genotypes can be indirectly characterized, e.g., using markers and/or directly characterized by nucleic acid sequencing.
  • germplasm refers to genetic material of or from an individual (e.g., a plant), a group of individuals (e.g., a plant line, variety or family), or a clone derived from a line, variety, species, or culture.
  • the germplasm can be part of an organism or cell, or can be separate from the organism or cell.
  • germplasm provides genetic material with a specific genetic makeup that provides a foundation for some or all of the hereditary qualities of an organism or cell culture.
  • germplasm includes cells, seed or tissues from which new plants may be grown, as well as plant parts that can be cultured into a whole plant (e.g., leaves, stems, buds, roots, pollen, cells, etc.).
  • cultivar and “variety” refer to a group of similar plants that by structural or genetic features and/or performance can be distinguished from other varieties within the same species.
  • exotic refers to any plant, line or germplasm that is not elite.
  • exotic plants/germplasms are not derived from any known elite plant or germplasm, but rather are selected to introduce one or more desired genetic elements into a breeding program (e.g., to introduce novel alleles into a breeding program).
  • hybrid in the context of plant breeding refers to a plant that is the offspring of genetically dissimilar parents produced by crossing plants of different lines or breeds or species, including but not limited to the cross between two inbred lines.
  • the term "inbred” refers to a substantially homozygous plant or variety.
  • the term may refer to a plant or plant variety that is substantially homozygous throughout the entire genome or that is substantially homozygous with respect to a portion of the genome that is of particular interest.
  • haplotype is the genotype of an individual at a plurality of genetic loci, i.e., a combination of alleles. Typically, the genetic loci that define a haplotype are physically and genetically linked, i.e., on the same chromosome segment.
  • haplotype can refer to polymorphisms at a particular locus, such as a single marker locus, or polymorphisms at multiple loci along a chromosomal segment.
  • heterologous refers to a nucleotide/polypeptide that originates from a foreign species, or, if from the same species, is substantially modified from its native form in composition and/or genomic locus by deliberate human intervention.
  • a plant in which at least one orthologous IPA1 gene encoding an SPL transcription factor is modified as described herein may have improved yield traits as compared to a plant that does not comprise the modification in the at least one orthologous IPA1 gene.
  • improved yield traits refers to any plant trait associated with growth, for example, biomass, yield, nitrogen use efficiency (NUE), inflorescence size/weight, fruit yield, fruit quality, fruit size, seed size, seed number, foliar tissue weight, nodulation number, nodulation mass, nodulation activity, number of seed heads, number of tillers, number of branches, number of flowers, number of tubers, tuber mass, bulb mass, number of seeds, total seed mass, rate of leaf emergence, rate of tiller/branch emergence, rate of seedling emergence, length of roots, number of roots, size and/or weight of root mass, or any combination thereof.
  • NUE nitrogen use efficiency
  • "improved yield traits” may include, but is not limited to, increased inflorescence production, increased fruit production (e.g., increased number, weight and/or size of fruit; e.g., increase number, weight, and/or size of ears for, e.g., maize), increased fruit quality, increased number, size and/or weight of roots, increased meristem size, increased seed size, increased biomass, increased leaf size, increased nitrogen use efficiency, increased height, increased intemode number and/or increased intemode length as compared to a control plant or part thereof (e.g., a plant that does not comprise a mutated endogenous IPA1 nucleic acid (e.g., a mutated IPA1 gene)).
  • a control plant or part thereof e.g., a plant that does not comprise a mutated endogenous IPA1 nucleic acid (e.g., a mutated IPA1 gene)
  • Improved yield traits can also result from increased planting density of plants of the invention.
  • a plant of the invention is capable of being planted at an increased density (as a consequence of altered plant architecture resulting from the endogenous mutation), which results in improved yield traits as compared to a control plant that is planted at the same density.
  • improved yield traits can be expressed as quantity of grain produced per area of land (e.g., bushels per acre of land).
  • control plant means a plant that does not contain an edited IPA1 gene or genes as described herein that imparts an enhanced/improved trait (e.g., yield trait) or altered phenotype.
  • a control plant is used to identify and select a plant edited as described herein and that has an enhanced trait or altered phenotype as compared to the control plant.
  • a suitable control plant can be a plant of the parental line used to generate a plant comprising a mutated IP A I gene(s), for example, a wild type plant devoid of an edit in an endogenous IPA1 gene as described herein.
  • a suitable control plant can also be a plant that contains recombinant nucleic acids that impart other traits, for example, a transgenic plant having enhanced herbicide tolerance.
  • a suitable control plant can in some cases be a progeny of a heterozygous or hemizygous transgenic plant line that is devoid of the mutated IPA1 gene as described herein, known as a negative segregant, or a negative isogenic line.
  • An enhanced trait may be, for example, decreased days from planting to maturity, increased stalk size, increased number of leaves, increased plant height growth rate in vegetative stage, increased ear size, increased ear dry weight per plant, increased number of kernels per ear, increased weight per kernel, increased number of kernels per plant, decreased ear void, extended grain fill period, reduced plant height, increased number of root branches, increased total root length, increased yield, increased nitrogen use efficiency, and increased water use efficiency as compared to a control plant.
  • An altered phenotype may be, for example, plant height, biomass, canopy area, anthocyanin content, chlorophyll content, water applied, water content, and water use efficiency.
  • a "trait” is a physiological, morphological, biochemical, or physical characteristic of a plant or particular plant material or cell. In some instances, this characteristic is visible to the human eye and can be measured mechanically, such as seed or plant size, weight, shape, form, length, height, growth rate and development stage, or can be measured by biochemical techniques, such as detecting the protein, starch, certain metabolites, or oil content of seed or leaves, or by observation of a metabolic or physiological process, for example, by measuring tolerance to water deprivation or particular salt or sugar concentrations, or by the measurement of the expression level of a gene or genes, for example, by employing Northern analysis, RT-PCR, microarray gene expression assays, or reporter gene expression systems, or by agricultural observations such as hyperosmotic stress tolerance or yield.
  • any technique can be used to measure the amount of, the comparative level of, or the difference in any selected chemical compound or macromolecule in the transgenic plants.
  • an "enhanced trait” means a characteristic of a plant resulting from mutations in mIPAl gene(s) as described herein.
  • Such traits include, but are not limited to, an enhanced agronomic trait characterized by enhanced plant morphology, physiology, growth and development, yield, nutritional enhancement, disease or pest resistance, or environmental or chemical tolerance.
  • an enhanced trait/altered phenotype may be, for example, decreased days from planting to maturity, increased stalk size, increased number of leaves, increased plant height growth rate in vegetative stage, increased ear size, increased ear dry weight per plant, increased number of kernels per ear, increased weight per kernel, increased number of kernels per plant, decreased ear void, extended grain fill period, reduced plant height, increased number of root branches, increased total root length, drought tolerance, increased water use efficiency, cold tolerance, increased nitrogen use efficiency, and increased yield.
  • a trait is increased yield under nonstress conditions or increased yield under environmental stress conditions.
  • Stress conditions can include both biotic and abiotic stress, for example, drought, shade, fungal disease, viral disease, bacterial disease, insect infestation, nematode infestation, cold temperature exposure, heat exposure, osmotic stress, reduced nitrogen nutrient availability, reduced phosphorus nutrient availability and high plant density.
  • biotic and abiotic stress for example, drought, shade, fungal disease, viral disease, bacterial disease, insect infestation, nematode infestation, cold temperature exposure, heat exposure, osmotic stress, reduced nitrogen nutrient availability, reduced phosphorus nutrient availability and high plant density.
  • Yield can be affected by many properties including without limitation, plant height, plant biomass, pod number, pod position on the plant, number of intemodes, incidence of pod shatter, grain size, ear size, ear tip filling, kernel abortion, efficiency of nodulation and nitrogen fixation, efficiency of nutrient assimilation, resistance to biotic and abiotic stress, carbon assimilation, plant architecture, resistance to lodging, percent seed germination, seedling vigor, and juvenile traits.
  • Yield can also be affected by efficiency of germination (including germination in stressed conditions), growth rate (including growth rate in stressed conditions), flowering time and duration, ear number, ear size, ear weight, seed number per ear or pod, seed size, composition of seed (starch, oil, protein) and characteristics of seed fill.
  • the term "trait modification” encompasses altering the naturally occurring trait by producing a detectable difference in a characteristic in a plant comprising a mutation in an endogenous IPA1 gene encoding an SPL transcription factor as described herein relative to a plant not comprising the mutation, such as a wild-type plant, or a negative segregant.
  • the trait modification can be evaluated quantitatively.
  • the trait modification can entail an increase or decrease in an observed trait characteristics or phenotype as compared to a control plant. It is known that there can be natural variations in a modified trait. Therefore, the trait modification observed entails a change of the normal distribution and magnitude of the trait characteristics or phenotype in the plants as compared to a control plant.
  • the present disclosure relates to a plant with improved economically important characteristics, more specifically increased yield. More specifically the present disclosure relates to a plant comprising a mutation(s) in an IPA1 gene(s) as described herein, wherein the plant has increased yield as compared to a control plant devoid of said mutation(s).
  • plants produced as described herein exhibit increased yield or improved yield trait components as compared to a control plant.
  • a plant of the present disclosure exhibits an improved trait that is related to yield, including but not limited to increased nitrogen use efficiency, increased nitrogen stress tolerance, increased water use efficiency and increased drought tolerance, as defined and discussed infra.
  • Yield can be defined as the measurable produce of economic value from a crop. Yield can be defined in the scope of quantity and/or quality. Yield can be directly dependent on several factors, for example, the number and size of organs, plant architecture (such as the number of branches, plant biomass, e.g., increased root biomass, steeper root angle and/or longer roots, and the like), flowering time and duration, grain fill period. Root architecture and development, photosynthetic efficiency, nutrient uptake, stress tolerance, early vigor, delayed senescence and functional stay green phenotypes may be factors in determining yield.
  • Reference herein to an increase/improvement in yield-related traits can also be taken to mean an increase in biomass (weight) of one or more parts of a plant, which can include above ground and/or below ground (harvestable) plant parts.
  • harvestable parts are seeds
  • performance of the methods of the disclosure results in plants with increased yield and in particular increased seed yield relative to the seed yield of suitable control plants.
  • the term "yield" of a plant can relate to vegetative biomass (root and/or shoot biomass), to reproductive organs, and/or to propagules (such as seeds) of that plant.
  • Increased yield of a plant of the present disclosure can be measured in a number of ways, including test weight, seed number per plant, seed weight, seed number per unit area (for example, seeds, or weight of seeds, per acre), bushels per acre, tons per acre, or kilo per hectare. Increased yield can result from improved utilization of key biochemical compounds, such as nitrogen, phosphorous and carbohydrate, or from improved responses to environmental stresses, such as cold, heat, drought, salt, shade, high plant density, and attack by pests or pathogens.
  • “Increased yield” can manifest as one or more of the following: (i) increased plant biomass (weight) of one or more parts of a plant, particularly aboveground (harvestable) parts, of a plant, increased root biomass (increased number of roots, increased root thickness, increased root length) or increased biomass of any other harvestable part; or (ii) increased early vigor, defined herein as an improved seedling aboveground area approximately three weeks post-germination.
  • Early vigor refers to active healthy plant growth especially during early stages of plant growth, and can result from increased plant fitness due to, for example, the plants being better adapted to their environment (for example, optimizing the use of energy resources, uptake of nutrients and partitioning carbon allocation between shoot and root).
  • Early vigor for example, can be a combination of the ability of seeds to germinate and emerge after planting and the ability of the young plants to grow and develop after emergence. Plants having early vigor also show increased seedling survival and better establishment of the crop, which often results in highly uniform fields with the majority of the plants reaching the various stages of development at substantially the same time, which often results in increased yield. Therefore, early vigor can be determined by measuring various factors, such as kernel weight, percentage germination, percentage emergence, seedling growth, seedling height, root length, root and shoot biomass, canopy size and color and others.
  • increased yield can also manifest as increased total seed yield, which may result from one or more of an increase in seed biomass (seed weight) due to an increase in the seed weight on a per plant and/or on an individual seed basis an increased number of, for example, flowers/panicles per plant; an increased number of pods; an increased number of nodes; an increased number of flowers ("florets") per panicle/plant; increased seed fill rate; an increased number of filled seeds; increased seed size (length, width, area, perimeter), which can also influence the composition of seeds; and/or increased seed volume, which can also influence the composition of seeds.
  • increased yield can be increased seed yield, for example, increased seed weight; increased number of filled seeds; and increased harvest index.
  • Increased yield can also result in modified architecture, or can occur because of modified plant architecture.
  • Increased yield can also manifest as increased harvest index, which is expressed as a ratio of the yield of harvestable parts, such as seeds, over the total biomass
  • the disclosure also extends to harvestable parts of a plant such as, but not limited to, seeds, leaves, fruits, flowers, bolls, pods, siliques, nuts, stems, rhizomes, tubers and bulbs.
  • the disclosure furthermore relates to products derived from a harvestable part of such a plant, such as dry pellets, powders, oil, fat and fatty acids, starch or proteins.
  • the present disclosure provides a method for increasing "yield" of a plant or "broad acre yield” of a plant or plant part defined as the harvestable plant parts per unit area, for example seeds, or weight of seeds, per acre, pounds per acre, bushels per acre, tones per acre, tons per acre, kilo per hectare.
  • nitrogen use efficiency refers to the processes which lead to an increase in the plant's yield, biomass, vigor, and growth rate per nitrogen unit applied.
  • the processes can include the uptake, assimilation, accumulation, signaling, sensing, retranslocation (within the plant) and use of nitrogen by the plant.
  • increased nitrogen use efficiency refers to the ability of plants to grow, develop, or yield faster or better than normal when subjected to the same amount of available/ applied nitrogen as under normal or standard conditions; ability of plants to grow, develop, or yield normally, or grow, develop, or yield faster or better when subjected to less than optimal amounts of available/ applied nitrogen, or under nitrogen limiting conditions.
  • nitrogen limiting conditions refers to growth conditions or environments that provide less than optimal amounts of nitrogen needed for adequate or successful plant metabolism, growth, reproductive success and/or viability.
  • the "increased nitrogen stress tolerance” refers to the ability of plants to grow, develop, or yield normally, or grow, develop, or yield faster or better when subjected to less than optimal amounts of available/applied nitrogen, or under nitrogen limiting conditions.
  • Increased plant nitrogen use efficiency can be translated in the field into either harvesting similar quantities of yield, while supplying less nitrogen, or increased yield gained by supplying optimal/sufficient amounts of nitrogen.
  • the increased nitrogen use efficiency can improve plant nitrogen stress tolerance and can also improve crop quality and biochemical constituents of the seed such as protein yield and oil yield.
  • the terms "increased nitrogen use efficiency”, “enhanced nitrogen use efficiency”, and “nitrogen stress tolerance” are used inter-changeably in the present disclosure to refer to plants with improved productivity under nitrogen limiting conditions.
  • water use efficiency refers to the amount of carbon dioxide assimilated by leaves per unit of water vapor transpired. It constitutes one of the most important traits controlling plant productivity in dry environments.
  • “Drought tolerance” refers to the degree to which a plant is adapted to arid or drought conditions. The physiological responses of plants to a deficit of water include leaf wilting, a reduction in leaf area, leaf abscission, and the stimulation of root growth by directing nutrients to the underground parts of the plants. Typically, plants are more susceptible to drought during flowering and seed development (the reproductive stages), as plant's resources are deviated to support root growth.
  • abscisic acid a plant stress hormone, induces the closure of leaf stomata (microscopic pores involved in gas exchange), thereby reducing water loss through transpiration, and decreasing the rate of photosynthesis. These responses improve the water-use efficiency of the plant on the short term.
  • ABA abscisic acid
  • the terms “increased water use efficiency”, “enhanced water use efficiency”, and “increased drought tolerance” are used inter-changeably in the present disclosure to refer to plants with improved productivity under water-limiting conditions.
  • increased water use efficiency refers to the ability of plants to grow, develop, or yield faster or better than normal when subjected to the same amount of available/ applied water as under normal or standard conditions; ability of plants to grow, develop, or yield normally, or grow, develop, or yield faster or better when subjected to reduced amounts of available/applied water (water input) or under conditions of water stress or water deficit stress.
  • increased drought tolerance refers to the ability of plants to grow, develop, or yield normally, or grow, develop, or yield faster or better than normal when subjected to reduced amounts of available/applied water and/or under conditions of acute or chronic drought; ability of plants to grow, develop, or yield normally when subjected to reduced amounts of available/applied water (water input) or under conditions of water deficit stress or under conditions of acute or chronic drought.
  • dwell stress refers to a period of dryness (acute or chronic/prolonged) that results in water deficit and subjects plants to stress and/or damage to plant tissues and/or negatively affects grain/crop yield; a period of dryness (acute or chronic/prolonged) that results in water deficit and/or higher temperatures and subjects plants to stress and/or damage to plant tissues and/or negatively affects grain/crop yield.
  • water deficit refers to the conditions or environments that provide less than optimal amounts of water needed for adequate/successful growth and development of plants.
  • water stress refers to the conditions or environments that provide improper (either less/insufficient or more/excessive) amounts of water than that needed for adequate/successful growth and development of plants/crops thereby subjecting the plants to stress and/or damage to plant tissues and/or negatively affecting grain/crop yield.
  • water deficit stress refers to the conditions or environments that provide less/insufficient amounts of water than that needed for adequate/successful growth and development of plants/crops thereby subjecting the plants to stress and/or damage to plant tissues and/or negatively affecting grain yield.
  • nucleic acid refers to RNA or DNA that is linear or branched, single or double stranded, or a hybrid thereof. The term also encompasses RNA/DNA hybrids.
  • dsRNA is produced synthetically, less common bases, such as inosine, 5-methylcytosine, 6-methyladenine, hypoxanthine and others can also be used for antisense, dsRNA, and ribozyme pairing.
  • polynucleotides that contain C-5 propyne analogues of uridine and cytidine have been shown to bind RNA with high affinity and to be potent antisense inhibitors of gene expression.
  • Other modifications, such as modification to the phosphodiester backbone, or the 2'-hydroxy in the ribose sugar group of the RNA can also be made.
  • nucleotide sequence refers to a heteropolymer of nucleotides or the sequence of these nucleotides from the 5' to 3' end of a nucleic acid molecule and includes DNA or RNA molecules, including cDNA, a DNA fragment or portion, genomic DNA, synthetic (e.g., chemically synthesized) DNA, plasmid DNA, mRNA, and anti-sense RNA, any of which can be single stranded or double stranded.
  • nucleic acid sequence “nucleic acid,” “nucleic acid molecule,” “nucleic acid construct,” “oligonucleotide” and “polynucleotide” are also used interchangeably herein to refer to a heteropolymer of nucleotides.
  • Nucleic acid molecules and/or nucleotide sequences provided herein are presented herein in the 5' to 3' direction, from left to right and are represented using the standard code for representing the nucleotide characters as set forth in the U.S. sequence rules, 37 CFR ⁇ 1.821 - 1.825 and the World Intellectual Property Organization (WIPO) Standard ST.25.
  • a "5' region” as used herein can mean the region of a polynucleotide that is nearest the 5' end of the polynucleotide.
  • an element in the 5' region of a polynucleotide can be located anywhere from the first nucleotide located at the 5' end of the polynucleotide to the nucleotide located halfway through the polynucleotide.
  • a "3' region” as used herein can mean the region of a polynucleotide that is nearest the 3' end of the polynucleotide.
  • an element in the 3' region of a polynucleotide can be located anywhere from the first nucleotide located at the 3' end of the polynucleotide to the nucleotide located halfway through the polynucleotide.
  • fragment refers to a nucleic acid that is reduced in length relative (e.g., reduced by 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12,
  • nucleic acid 450, 500, 550, 600, 650, 700, 750, 800, 850, or 900 or more nucleotides or any range or value therein) to a reference nucleic acid and that comprises, consists essentially of and/or consists of a nucleotide sequence of contiguous nucleotides identical or almost identical (e.g., 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%,
  • nucleic acid fragment may be, where appropriate, included in a larger polynucleotide of which it is a constituent.
  • a repeat sequence of guide nucleic acid of this invention may comprise a "portion" of a wild type CRISPR-Cas repeat sequence (e.g., a wild Type CRISR-Cas repeat, e.g., a repeat from the CRISPR Cas system of, for example, a Cas9, Casl2a (Cpfl), Casl2b, Casl2c (C2c3), Casl2d (CasY), Casl2e (CasX), Casl2g, Casl2h, Casl2i, C2c4, C2c5, C2c8, C2c9, C2cl0, Casl4a, Casl4b, and/or a Casl4c, and the like).
  • a wild type CRISPR-Cas repeat sequence e.g., a wild Type CRISR-Cas repeat, e.g., a wild Type CRISR-Cas repeat, e.g
  • a fragment or a portion of a CRN nucleic acid may comprise, consist essentially of or consist of about 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 120, 130, 140,
  • 1100, 1120, 1140, 1160, 1180, 1200, 1220, 1240, 1260, or 1280 consecutive nucleotides or any range or value therein of a nucleic acid encoding a CRN polypeptide, optionally a fragment or a portion of a CRN gene may be about 10, 20, 30, 40, 50, 90, 100 to about 200, 300, 400, 500,
  • sequence-specific nucleic acid binding domain e.g., sequence-specific DNA binding domain, sequence-specific RNA binding domain
  • a "sequence-specific nucleic acid binding domain” may bind to one or more fragments or portions of nucleotide sequences encoding CRN polypeptides as described herein.
  • fragment may refer to a polypeptide that is reduced in length relative to a reference polypeptide and that comprises, consists essentially of and/or consists of an amino acid sequence of contiguous amino acids identical or almost identical (e.g., 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% identical) to a corresponding portion of the reference polypeptide.
  • a polypeptide fragment may be, where appropriate, included in a larger polypeptide of which it is a constituent.
  • the polypeptide fragment comprises, consists essentially of or consists of at least about 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80,
  • a fragment or portion of a CRN polypeptide may comprise, consist essentially of or consist of about 10, 11, 12, 13, 14, 15, 16,
  • the term "functional fragment” refers to nucleic acid that encodes a functional fragment of a polypeptide.
  • gene refers to a nucleic acid molecule capable of being used to produce mRNA, antisense RNA, miRNA, anti-microRNA antisense oligodeoxyribonucleotide (AMO) and the like. Genes may or may not be capable of being used to produce a functional protein or gene product. Genes can include both coding and non-coding regions (e.g., introns, regulatory elements, promoters, enhancers, termination sequences and/or 5' and 3' untranslated regions).
  • a gene may be "isolated” by which is meant a nucleic acid that is substantially or essentially free from components normally found in association with the nucleic acid in its natural state. Such components include other cellular material, culture medium from recombinant production, and/or various chemicals used in chemically synthesizing the nucleic acid.
  • mutant refers to point mutations (e.g., missense, or nonsense, or insertions or deletions of single base pairs that result in frame shifts), insertions, deletions, and/or truncations.
  • mutations are typically described by identifying the original residue followed by the position of the residue within the sequence and by the identity of the newly substituted residue.
  • a truncation can include a truncation at the C-terminal end of a polypeptide or at the N-terminal end of a polypeptide.
  • a truncation of a polypeptide can be the result of a deletion of the corresponding 5' end or 3' end of the gene encoding the polypeptide.
  • a frameshift mutation can occur when deletions or insertions of one or more base pairs are introduced into a gene. Frameshift mutations in a gene can result in the production of a polypeptide that is longer, shorter or the same length as the wild type polypeptide depending on when the first stop codon occurs following the mutated region of the gene.
  • a deletion useful with this invention may result in an in-frame mutation.
  • such a deletion may be a dominant negative mutation, a semi dominant mutation, a hypomorphic mutation, a weak loss-of-function mutation or a null allele, which when comprised in a plant can result in the plant exhibiting increased kernel number as compared to a plant not comprising said deletion.
  • a deletion may be a deletion of 1 base pair to about 160 base pairs (e.g., about 3 base pairs to about 10 base pairs, about 3 base pairs to about 20 base pairs, about 3 base pairs to about 30 base pairs, about 3 base pairs to about 35 base pairs, about 3 base pairs to about 40 base pairs, about 3 base pairs to about 45 base pairs, about 3 base pairs to about 50 base pairs, about 3 base pairs to about 80 base pairs, about 3 base pairs to about 100 base pairs, about 3 base pairs to about 160 base pairs, about 10 base pairs to about 160 base pairs, about 15 base pairs to about 160 base pairs, about 20 base pairs to about 160 base pairs, about 30 base pairs to about 160 base pairs, about 50 base pairs to about 160 base pairs, about 75 base pairs to about 160 base pairs, or about 96 base pairs to about 160 base pairs; e.g., about 1, 2,3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 base pairs to about 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100
  • An CRN gene may be edited in more than one location, thereby providing a CRN gene comprising more than one mutation.
  • a plant may also exhibit larger meristems, maintenance of root meristems and increased yield and increased disease resistance. Additional advantages of the present invention include, but are not limited to regulating growth, regulating meristem size, regulating vascular development, controlling organ number, facilitating increased plant regeneration from tissue culture, increasing fruit size and or controlling plant secondary growth.
  • complementarity refers to the natural binding of polynucleotides under permissive salt and temperature conditions by base-pairing.
  • sequence "A-G-T” (5' to 3') binds to the complementary sequence "T-C-A" (3' to 5').
  • Complementarity between two single-stranded molecules may be "partial,” in which only some of the nucleotides bind, or it may be complete when total complementarity exists between the single stranded molecules.
  • the degree of complementarity between nucleic acid strands has significant effects on the efficiency and strength of hybridization between nucleic acid strands.
  • “Complement,” as used herein, can mean 100% complementarity with the comparator nucleotide sequence or it can mean less than 100% complementarity (e.g., about 70%, 71%,
  • homologues Different nucleic acids or proteins having homology are referred to herein as "homologues.”
  • the term homologue includes homologous sequences from the same and from other species and orthologous sequences from the same and other species.
  • “Homology” refers to the level of similarity between two or more nucleic acid and/or amino acid sequences in terms of percent of positional identity (i.e., sequence similarity or identity). Homology also refers to the concept of similar functional properties among different nucleic acids or proteins.
  • the compositions and methods of the invention further comprise homologues to the nucleotide sequences and polypeptide sequences of this invention.
  • Orthologous refers to homologous nucleotide sequences and / or amino acid sequences in different species that arose from a common ancestral gene during speciation.
  • a homologue of a nucleotide sequence of this invention has a substantial sequence identity (e.g., at least about 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or 100%) to said nucleotide sequence of the invention.
  • sequence identity refers to the extent to which two optimally aligned polynucleotide or polypeptide sequences are invariant throughout a window of alignment of components, e.g., nucleotides or amino acids. "Identity” can be readily calculated by known methods including, but not limited to, those described in: Computational Molecular Biology (Lesk, A. M., ed.) Oxford University Press, New York (1988); Biocomputing: Informatics and Genome Projects (Smith, D. W., ed.) Academic Press, New York (1993); Computer Analysis of Sequence Data, Part I (Griffin, A. M., and Griffin, H.
  • percent sequence identity refers to the percentage of identical nucleotides in a linear polynucleotide sequence of a reference (“query”) polynucleotide molecule (or its complementary strand) as compared to a test ("subject") polynucleotide molecule (or its complementary strand) when the two sequences are optimally aligned.
  • percent sequence identity can refer to the percentage of identical amino acids in an amino acid sequence as compared to a reference polypeptide.
  • the phrase "substantially identical,” or “substantial identity” in the context of two nucleic acid molecules, nucleotide sequences, or polypeptide sequences refers to two or more sequences or subsequences that have at least about 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or 100% nucleotide or amino acid residue identity, when compared and aligned for maximum correspondence, as measured using one of the following sequence comparison algorithms or by visual inspection.
  • the substantial identity exists over a region of consecutive nucleotides of a nucleotide sequence of the invention that is about 10 nucleotides to about 20 nucleotides, about 10 nucleotides to about 25 nucleotides, about 10 nucleotides to about 30 nucleotides, about 15 nucleotides to about 25 nucleotides, about 30 nucleotides to about 40 nucleotides, about 50 nucleotides to about 60 nucleotides, about 70 nucleotides to about 80 nucleotides, about 90 nucleotides to about 100 nucleotides, about 100 nucleotides to about 200 nucleotides, about 100 nucleotides to about 300 nucleotides, about 100 nucleotides to about 400 nucleotides, about 100 nucleotides to about 500 nucleotides, about 100 nucleotides to about 600 nucleotides, about 100 nucleotides to about 800
  • nucleotide sequences can be substantially identical over at least about 20 consecutive nucleotides (e.g., about 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 50, 60, 70, or 80 or more consecutive nucleotides).
  • the substantial identity exists over a region of consecutive amino acid residues of a polypeptide of the invention that is about 3 amino acid residues to about 20 amino acid residues, about 5 amino acid residues to about 25 amino acid residues, about 7 amino acid residues to about 30 amino acid residues, about 10 amino acid residues to about 25 amino acid residues, about 15 amino acid residues to about 30 amino acid residues, about 20 amino acid residues to about 40 amino acid residues, about 25 amino acid residues to about 40 amino acid residues, about 25 amino acid residues to about 50 amino acid residues, about 30 amino acid residues to about 50 amino acid residues, about 40 amino acid residues to about 50 amino acid residues, about 40 amino acid residues to about 50 amino acid residues, about 40 amino acid residues to about 70 amino acid residues, about 50 amino acid residues to about 70 amino acid residues, about 60 amino acid residues to about 80 amino acid residues, about 70 amino acid residues to about 80 amino acid residues, about 90 amino acid residues to about 100 amino acid residues, or more amino acid residue
  • polypeptide sequences can be substantially identical to one another over at least about 8 consecutive amino acid residues (e.g., about 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45,
  • two or more CRN polypeptides may be identical or substantially identical (e.g., at least 70% to 99.9% identical, e.g., about 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%,
  • sequence comparison typically one sequence acts as a reference sequence to which test sequences are compared.
  • test and reference sequences are entered into a computer, subsequence coordinates are designated if necessary, and sequence algorithm program parameters are designated.
  • sequence comparison algorithm then calculates the percent sequence identity for the test sequence(s) relative to the reference sequence, based on the designated program parameters.
  • Optimal alignment of sequences for aligning a comparison window are well known to those skilled in the art and may be conducted by tools such as the local homology algorithm of Smith and Waterman, the homology alignment algorithm of Needleman and Wunsch, the search for similarity method of Pearson and Lipman, and optionally by computerized implementations of these algorithms such as GAP, BESTFIT, FASTA, and TFASTA available as part of the GCG® Wisconsin Package® (Accelrys Inc., San Diego, CA).
  • An "identity fraction" for aligned segments of a test sequence and a reference sequence is the number of identical components which are shared by the two aligned sequences divided by the total number of components in the reference sequence segment, e.g., the entire reference sequence or a smaller defined part of the reference sequence.
  • Percent sequence identity is represented as the identity fraction multiplied by 100.
  • the comparison of one or more polynucleotide sequences may be to a full-length polynucleotide sequence or a portion thereof, or to a longer polynucleotide sequence.
  • percent identity may also be determined using BLASTX version 2.0 for translated nucleotide sequences and BLASTN version 2.0 for polynucleotide sequences.
  • Two nucleotide sequences may also be considered substantially complementary when the two sequences hybridize to each other under stringent conditions.
  • two nucleotide sequences considered to be substantially complementary hybridize to each other under highly stringent conditions.
  • Stringent hybridization conditions and “stringent hybridization wash conditions” in the context of nucleic acid hybridization experiments such as Southern and Northern hybridizations are sequence dependent and are different under different environmental parameters. An extensive guide to the hybridization of nucleic acids is found in Tijssen Laboratory Techniques in Biochemistry and Molecular Biology-Hybridization with Nucleic Acid Probes part I chapter 2 "Overview of principles of hybridization and the strategy of nucleic acid probe assays" Elsevier, New York (1993).
  • highly stringent hybridization and wash conditions are selected to be about 5°C lower than the thermal melting point (T m ) for the specific sequence at a defined ionic strength and pH.
  • the Tm is the temperature (under defined ionic strength and pH) at which 50% of the target sequence hybridizes to a perfectly matched probe.
  • Very stringent conditions are selected to be equal to the T m for a particular probe.
  • An example of stringent hybridization conditions for hybridization of complementary nucleotide sequences which have more than 100 complementary residues on a filter in a Southern or northern blot is 50% formamide with 1 mg of heparin at 42°C, with the hybridization being carried out overnight.
  • An example of highly stringent wash conditions is 0.1 5M NaCl at 72°C for about 15 minutes.
  • An example of stringent wash conditions is a 0.2x SSC wash at 65°C for 15 minutes (see, Sambrook, infra, for a description of SSC buffer).
  • a high stringency wash is preceded by a low stringency wash to remove background probe signal.
  • An example of a medium stringency wash for a duplex of, e.g., more than 100 nucleotides, is lx SSC at 45°C for 15 minutes.
  • An example of a low stringency wash for a duplex of, e.g., more than 100 nucleotides, is 4-6x SSC at 40°C for 15 minutes.
  • stringent conditions typically involve salt concentrations of less than about 1.0 M Na ion, typically about 0.01 to 1.0 M Na ion concentration (or other salts) at pH 7.0 to 8.3, and the temperature is typically at least about 30°C.
  • Stringent conditions can also be achieved with the addition of destabilizing agents such as formamide.
  • a signal to noise ratio of 2x (or higher) than that observed for an unrelated probe in the particular hybridization assay indicates detection of a specific hybridization.
  • Nucleotide sequences that do not hybridize to each other under stringent conditions are still substantially identical if the proteins that they encode are substantially identical. This can occur, for example, when a copy of a nucleotide sequence is created using the maximum codon degeneracy permitted by the genetic code.
  • a polynucleotide and/or recombinant nucleic acid construct of this invention may be codon optimized for expression.
  • the polynucleotides, nucleic acid constructs, expression cassettes, and/or vectors of the editing systems of the invention e.g., comprising/encoding a sequence-specific nucleic acid binding domain (e.g., a sequence-specific nucleic acid binding domain from a polynucleotide-guided endonuclease, a zinc finger nuclease, a transcription activator-like effector nuclease (TALEN), an Argonaute protein, and/or a CRISPR-Cas endonuclease (e.g., CRISPR-Cas effector protein) (e.g., a Type I CRISPR-Cas effector protein, a Type II CRISPR- Cas effector protein, a Type III C
  • the codon optimized nucleic acids, polynucleotides, expression cassettes, and/or vectors of the invention have about 70% to about 99.9% (e.g., 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%. 99.9% or 100%) identity or more to the reference nucleic acids, polynucleotides, expression cassettes, and/or vectors that have not been codon optimized.
  • a polynucleotide or nucleic acid construct of the invention may be operatively associated with a variety of promoters and/or other regulatory elements for expression in a plant and/or a cell of a plant.
  • a polynucleotide or nucleic acid construct of this invention may further comprise one or more promoters, introns, enhancers, and/or terminators operably linked to one or more nucleotide sequences.
  • a promoter may be operably associated with an intron (e.g., Ubil promoter and intron).
  • a promoter associated with an intron maybe referred to as a "promoter region" (e.g., Ubil promoter and intron).
  • operably linked or “operably associated” as used herein in reference to polynucleotides, it is meant that the indicated elements are functionally related to each other and are also generally physically related.
  • operably linked refers to nucleotide sequences on a single nucleic acid molecule that are functionally associated.
  • a first nucleotide sequence that is operably linked to a second nucleotide sequence means a situation when the first nucleotide sequence is placed in a functional relationship with the second nucleotide sequence.
  • a promoter is operably associated with a nucleotide sequence if the promoter effects the transcription or expression of said nucleotide sequence.
  • control sequences e.g., promoter
  • the control sequences need not be contiguous with the nucleotide sequence to which it is operably associated, as long as the control sequences function to direct the expression thereof.
  • intervening untranslated, yet transcribed, nucleic acid sequences can be present between a promoter and the nucleotide sequence, and the promoter can still be considered "operably linked" to the nucleotide sequence.
  • polypeptides refers to the atachment of one polypeptide to another.
  • a polypeptide may be linked to another polypeptide (at the N- terminus or the C-terminus) directly (e.g., via a peptide bond) or through a linker.
  • linker refers to a chemical group, or a molecule linking two molecules or moieties, e.g., two domains of a fusion protein, such as, for example, a nucleic acid binding polypeptide or domain and peptide tag and/or a reverse transcriptase and an affinity polypeptide that binds to the peptide tag; or a DNA endonuclease polypeptide or domain and peptide tag and/or a reverse transcriptase and an affinity polypeptide that binds to the peptide tag.
  • a linker may be comprised of a single linking molecule or may comprise more than one linking molecule.
  • the linker can be an organic molecule, group, polymer, or chemical moiety such as a bivalent organic moiety.
  • the linker may be an amino acid, or it may be a peptide. In some embodiments, the linker is a peptide.
  • a peptide linker useful with this invention may be about 2 to about 100 or more amino acids in length, for example, about 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13,
  • amino acids in length e.g., about 2 to about 40, about 2 to about 50, about 2 to about 60, about 4 to about 40, about 4 to about 50, about 4 to about 60, about 5 to about 40, about 5 to about 50, about 5 to about 60, about 9 to about 40, about 9 to about 50, about 9 to about 60, about 10 to about 40, about 10 to about 50, about 10 to about 60, or about 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25 amino acids to about 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72,
  • a peptide linker may be a GS linker.
  • the term "linked,” or “fused" in reference to polynucleotides refers to the attachment of one polynucleotide to another.
  • two or more polynucleotide molecules may be linked by a linker that can be an organic molecule, group, polymer, or chemical moiety such as a bivalent organic moiety.
  • a polynucleotide may be linked or fused to another polynucleotide (at the 5' end or the 3' end) via a covalent or non-covenant linkage or binding, including e.g., Watson-Crick base-pairing, or through one or more linking nucleotides.
  • a polynucleotide motif of a certain structure may be inserted within another polynucleotide sequence (e.g. extension of the hairpin structure in the guide RNA).
  • the linking nucleotides may be naturally occurring nucleotides.
  • the linking nucleotides may be non-naturally occurring nucleotides.
  • a “promoter” is a nucleotide sequence that controls or regulates the transcription of a nucleotide sequence (e.g., a coding sequence) that is operably associated with the promoter.
  • the coding sequence controlled or regulated by a promoter may encode a polypeptide and/or a functional RNA.
  • a “promoter” refers to a nucleotide sequence that contains a binding site for RNA polymerase II and directs the initiation of transcription.
  • promoters are found 5', or upstream, relative to the start of the coding region of the corresponding coding sequence.
  • a promoter may comprise other elements that act as regulators of gene expression; e.g., a promoter region.
  • Promoters useful with this invention can include, for example, constitutive, inducible, temporally regulated, developmentally regulated, chemically regulated, tissue-preferred and/or tissue-specific promoters for use in the preparation of recombinant nucleic acid molecules, e.g., "synthetic nucleic acid constructs" or "protein-RNA complex.” These various types of promoters are known in the art.
  • promoter may vary depending on the temporal and spatial requirements for expression, and also may vary based on the host cell to be transformed. Promoters for many different organisms are well known in the art. Based on the extensive knowledge present in the art, the appropriate promoter can be selected for the particular host organism of interest. Thus, for example, much is known about promoters upstream of highly constitutively expressed genes in model organisms and such knowledge can be readily accessed and implemented in other systems as appropriate. In some embodiments, a promoter functional in a plant may be used with the constructs of this invention.
  • Non-limiting examples of a promoter useful for driving expression in a plant include the promoter of the RubisCo small subunit gene 1 (PrbcSl), the promoter of the actin gene (Pactin), the promoter of the nitrate reductase gene (Pnr) and the promoter of duplicated carbonic anhydrase gene 1 (Pdcal) (See, Walker et al. Plant Cell Rep. 23:727-735 (2005); Li et al. Gene 403:132-142 (2007); Li et al. Mol Biol. Rep. 37:1143-1154 (2010)). PrbcSl and Pactin are constitutive promoters and Pnr and Pdcal are inducible promoters.
  • PrbcSl and Pactin are constitutive promoters and Pnr and Pdcal are inducible promoters.
  • a promoter useful with this invention is RNA polymerase II (Pol II) promoter.
  • a U6 promoter or a 7SL promoter from Zea mays may be useful with constructs of this invention.
  • the U6c promoter and/or 7SL promoter from Zea mays may be useful for driving expression of a guide nucleic acid.
  • a U6c promoter, U6i promoter and/or 7SL promoter from Glycine max may be useful with constructs of this invention. In some embodiments, the U6c promoter, U6i promoter and/or 7SL promoter from Glycine max may be useful for driving expression of a guide nucleic acid.
  • constitutive promoters useful for plants include, but are not limited to, cestrum virus promoter (cmp) (U.S. Patent No. 7,166,770), the rice actin 1 promoter (Wang et al. (1992) Mol. Cell. Biol. 12:3399-3406; as well as US Patent No. 5,641,876), CaMV 35S promoter (Odell et al. (1985) Nature 313:810-812), CaMV 19S promoter (Lawton et al. (1987) Plant Mol. Biol. 9:315-324), nos promoter (Ebert et al. (1987) Proc. Natl. Acad.
  • the maize ubiquitin promoter (UbiP) has been developed in transgenic monocot systems and its sequence and vectors constructed for monocot transformation are disclosed in the patent publication EP 0342926.
  • the ubiquitin promoter is suitable for the expression of the nucleotide sequences of the invention in transgenic plants, especially monocotyledons.
  • the promoter expression cassettes described by McElroy et al. ⁇ Mol. Gen. Genet. 231: 150-160 (1991)) can be easily modified for the expression of the nucleotide sequences of the invention and are particularly suitable for use in monocotyledonous hosts.
  • tissue specific/tissue preferred promoters can be used for expression of a heterologous polynucleotide in a plant cell.
  • Tissue specific or preferred expression paterns include, but are not limited to, green tissue specific or preferred, root specific or preferred, stem specific or preferred, flower specific or preferred or pollen specific or preferred. Promoters suitable for expression in green tissue include many that regulate genes involved in photosynthesis and many of these have been cloned from both monocotyledons and dicotyledons.
  • a promoter useful with the invention is the maize PEPC promoter from the phosphoenol carboxylase gene (Hudspeth & Grula, Plant Molec. Biol. 12:579-589 (1989)).
  • tissue-specific promoters include those associated with genes encoding the seed storage proteins (such as b-conglycinin, cruciferin, napin and phaseolin), zein or oil body proteins (such as oleosin), or proteins involved in fatty acid biosynthesis (including acyl carrier protein, stearoyl-ACP desaturase and fatty acid desaturases (fad 2-1)), and other nucleic acids expressed during embryo development (such as Bce4, see, e.g., Kridl et al. (1991) SeedSci. Res. 1:209-219; as well as EP Patent No. 255378).
  • seed storage proteins such as b-conglycinin, cruciferin, napin and phaseolin
  • zein or oil body proteins such as oleosin
  • proteins involved in fatty acid biosynthesis including acyl carrier protein, stearoyl-ACP desaturase and fatty acid desaturases (fad 2-1)
  • Tissue-specific or tissue-preferential promoters useful for the expression of the nucleotide sequences of the invention in plants, particularly maize include but are not limited to those that direct expression in root, pith, leaf or pollen. Such promoters are disclosed, for example, in WO 93/07278, herein incorporated by reference in its entirety.
  • tissue specific or tissue preferred promoters useful with the invention the cotton rubisco promoter disclosed in US Patent 6,040,504; the rice sucrose synthase promoter disclosed in US Patent 5,604,121; the root specific promoter described by de Framond (FEBS 290:103-106 (1991); EP 0452269 to Ciba- Geigy); the stem specific promoter described in U.S.
  • Patent 5,625,136 (to Ciba-Geigy) and which drives expression of the maize trpA gene; the cestrum yellow leaf curling virus promoter disclosed in WO 01/73087; and pollen specific or preferred promoters including, but not limited to, ProOsLPSlO and ProOsLPSl 1 from rice (Nguyen et al. Plant Biotechnol. Reports 9(5):297-306 (2015)), ZmSTK2_USP from maize (Wang et al. Genome 60(6):485-495 (2017)), LAT52 and LAT59 from tomato (Twell et al. Development 109(3):705- 713 (1990)), Zml3 (U.S. Patent No. 10,421,972), PLA2-5 promoter from arabidopsis (U.S. Patent No. 7,141,424), and/or the ZmC5 promoter from maize (International PCT Publication No. WO 1999/042587.
  • plant tissue-specific/tissue preferred promoters include, but are not limited to, the root hair-specific cis-elements (RHEs) (Kim et al. The Plant Cell 18:2958- 2970 (2006)), the root-specific promoters RCc3 (Jeong et al. Plant Physiol. 153:185-197 (2010)) and RB7 (U.S. Patent No. 5459252), the lectin promoter (Lindstrom et al. (1990) Der. Genet. 11:160-167; and Vodkin (1983) Prog. Clin. Biol. Res. 138:87-98), com alcohol dehydrogenase 1 promoter (Dennis et al.
  • RHEs root hair-specific cis-elements
  • RuBP carboxylase promoter Ceashmore, "Nuclear genes encoding the small subunit of ribulose-l,5-bisphosphate carboxylase" pp. 29-39 In: Genetic Engineering of Plants (Hollaender ed., Plenum Press 1983; and Poulsen et al. (1986) Mol. Gen. Genet. 205:193-200), Ti plasmid mannopine synthase promoter (Langridge et al. (1989) Proc. Natl. Acad. Sci. USA 86:3219-3223), Ti plasmid nopaline synthase promoter (Langridge et al.
  • petunia chalcone isomerase promoter van Tunen et al. (1988) EMBOJ. 7:1257-1263
  • bean glycine rich protein 1 promoter Kerman et al. (1989) Genes Dev. 3:1639-1646
  • truncated CaMV 35S promoter O'Dell et al. (1985) Nature 313:810-812)
  • potato patatin promoter Wenzler et al. (1989) Plant Mol. Biol. 13:347-354
  • root cell promoter Yamamoto et al. (1990 ) Nucleic Acids Res . 18:7449
  • maize zein promoter Yama et al. (1987) Mol. Gen.
  • Useful for seed-specific expression is the pea vicilin promoter (Czako et al. (1992) Mol. Gen. Genet. 235:33-40; as well as the seed-specific promoters disclosed in U.S. Patent No. 5,625,136.
  • Useful promoters for expression in mature leaves are those that are switched at the onset of senescence, such as the SAG promoter from Arabidopsis (Gan et al. (1995) Science 270:1986-1988).
  • promoters functional in chloroplasts can be used.
  • Non-limiting examples of such promoters include the bacteriophage T3 gene 9 5' UTR and other promoters disclosed in U.S. Patent No. 7,579,516.
  • Other promoters useful with the invention include but are not limited to the S-E9 small subunit RuBP carboxylase promoter and the Kunitz trypsin inhibitor gene promoter (Kti3).
  • Additional regulatory elements useful with this invention include, but are not limited to, introns, enhancers, termination sequences and/or 5' and 3' untranslated regions.
  • An intron useful with this invention can be an intron identified in and isolated from a plant and then inserted into an expression cassette to be used in transformation of a plant.
  • introns can comprise the sequences required for self-excision and are incorporated into nucleic acid constructs/expression cassettes in frame.
  • An intron can be used either as a spacer to separate multiple protein-coding sequences in one nucleic acid construct, or an intron can be used inside one protein-coding sequence to, for example, stabilize the mRNA. If they are used within a protein-coding sequence, they are inserted "in-frame" with the excision sites included.
  • Introns may also be associated with promoters to improve or modify expression.
  • a promoter/intron combination useful with this invention includes but is not limited to that of the maize Ubil promoter and intron (see, e.g., SEQ ID NO:21 and SEQ ID NO:22).
  • Non-limiting examples of introns useful with the present invention include introns from the ADHI gene (e.g., Adhl-S introns 1, 2 and 6), the ubiquitin gene (Ubil), the RuBisCO small subunit (rbcS) gene, the RuBisCO large subunit (rbcL) gene, the actin gene (e.g., actin-1 intron), the pyruvate dehydrogenase kinase gene (pdk), the nitrate reductase gene (nr), the duplicated carbonic anhydrase gene 1 (Tdcal), the psbA gene, the atpA gene, or any combination thereof.
  • ADHI gene e.g., Adhl-S introns 1, 2 and 6
  • the ubiquitin gene Ubil
  • the RuBisCO small subunit (rbcS) gene the RuBisCO large subunit (rbcL) gene
  • the actin gene e.g., actin-1 in
  • a polynucleotide and/or a nucleic acid construct of the invention can be an "expression cassette" or can be comprised within an expression cassette.
  • expression cassette means a recombinant nucleic acid molecule comprising, for example, a one or more polynucleotides of the invention (e.g., a polynucleotide encoding a sequence-specific nucleic acid binding domain, a polynucleotide encoding a deaminase protein or domain, a polynucleotide encoding a reverse transcriptase protein or domain, a polynucleotide encoding a 5'-3' exonuclease polypeptide or domain, a guide nucleic acid and/or reverse transcriptase (RT) template), wherein polynucleotide(s) is/are operably associated with one or more control sequences (e.g., a promoter, terminator and
  • control sequences e.g
  • one or more expression cassettes may be provided, which are designed to express, for example, a nucleic acid construct of the invention (e.g., a polynucleotide encoding a sequence-specific nucleic acid binding domain (e.g., a sequence-specific DNA binding domain), a polynucleotide encoding a nuclease polypeptide/domain, a polynucleotide encoding a deaminase protein/domain, a polynucleotide encoding a reverse transcriptase protein/domain, a polynucleotide encoding a 5'-3' exonuclease polypeptide/domain, a polynucleotide encoding a peptide tag, and/or a polynucleotide encoding an affinity polypeptide, and the like, or comprising a guide nucleic acid, an extended guide nucleic acid, and/or RT template, and the
  • an expression cassette of the present invention comprises more than one polynucleotide
  • the polynucleotides may be operably linked to a single promoter that drives expression of all of the polynucleotides or the polynucleotides may be operably linked to one or more separate promoters (e.g., three polynucleotides may be driven by one, two or three promoters in any combination).
  • the promoters may be the same promoter, or they may be different promoters.
  • a polynucleotide encoding a sequence specific nucleic acid binding domain may each be operably linked to a single promoter, or separate promoters in any combination.
  • An expression cassette comprising a nucleic acid construct of the invention may be chimeric, meaning that at least one (e.g., one or more) of its components is heterologous with respect to at least one of its other components (e.g., a promoter from the host organism operably linked to a polynucleotide of interest to be expressed in the host organism, wherein the polynucleotide of interest is from a different organism than the host or is not normally found in association with that promoter).
  • An expression cassette may also be one that is naturally occurring but has been obtained in a recombinant form useful for heterologous expression.
  • An expression cassette can optionally include a transcriptional and/or translational termination region (i.e., termination region) and/or an enhancer region that is functional in the selected host cell.
  • a transcriptional and/or translational termination region i.e., termination region
  • an enhancer region that is functional in the selected host cell.
  • a variety of transcriptional terminators and enhancers are known in the art and are available for use in expression cassettes. Transcriptional terminators are responsible for the termination of transcription and correct mRNA polyadenylation.
  • a termination region and/or the enhancer region may be native to the transcriptional initiation region, may be native to, for example, a gene encoding a sequence-specific nucleic acid binding protein, a gene encoding a nuclease, a gene encoding a reverse transcriptase, a gene encoding a deaminase, and the like, or may be native to a host cell, or may be native to another source (e.g., foreign or heterologous to, for example, to a promoter, to a gene encoding a sequence-specific nucleic acid binding protein, a gene encoding a nuclease, a gene encoding a reverse transcriptase, a gene encoding a deaminase, and the like, or to the host cell, or any combination thereof).
  • An expression cassette of the invention also can include a polynucleotide encoding a selectable marker, which can be used to select a transformed host cell.
  • selectable marker means a polynucleotide sequence that when expressed imparts a distinct phenotype to the host cell expressing the marker and thus allows such transformed cells to be distinguished from those that do not have the marker.
  • Such a polynucleotide sequence may encode either a selectable or screenable marker, depending on whether the marker confers a trait that can be selected for by chemical means, such as by using a selective agent (e.g., an antibiotic and the like), or on whether the marker is simply a trait that one can identify through observation or testing, such as by screening (e.g., fluorescence).
  • a selective agent e.g., an antibiotic and the like
  • screening e.g., fluorescence
  • vectors refers to a composition for transferring, delivering or introducing a nucleic acid (or nucleic acids) into a cell.
  • a vector comprises a nucleic acid construct (e.g., expression cassette(s)) comprising the nucleotide sequence(s) to be transferred, delivered or introduced.
  • vectors for use in transformation of host organisms are well known in the art.
  • Non-limiting examples of general classes of vectors include viral vectors, plasmid vectors, phage vectors, phagemid vectors, cosmid vectors, fosmid vectors, bacteriophages, artificial chromosomes, minicircles, or Agrobacterium binary vectors in double or single stranded linear or circular form which may or may not be self-transmissible or mobilizable.
  • a viral vector can include, but is not limited, to a retroviral, lentiviral, adenoviral, adeno-associated, or herpes simplex viral vector.
  • a vector as defined herein can transform a prokaryotic or eukaryotic host either by integration into the cellular genome or exist extrachromosomally (e.g., autonomous replicating plasmid with an origin of replication).
  • shuttle vectors by which is meant a DNA vehicle capable, naturally or by design, of replication in two different host organisms, which may be selected from actinomycetes and related species, bacteria and eukaryotic (e.g., higher plant, mammalian, yeast or fungal cells).
  • the nucleic acid in the vector is under the control of, and operably linked to, an appropriate promoter or other regulatory elements for transcription in a host cell.
  • the vector may be a bi-functional expression vector which functions in multiple hosts.
  • nucleic acid or polynucleotide of this invention and/or expression cassettes comprising the same may be comprised in vectors as described herein and as known in the art.
  • contact refers to placing the components of a desired reaction together under conditions suitable for carrying out the desired reaction (e.g., transformation, transcriptional control, genome editing, nicking, and/or cleavage).
  • a target nucleic acid may be contacted with a sequence-specific nucleic acid binding protein (e.g., polynucleotide-guided endonuclease, a CRISPR-Cas endonuclease (e.g., CRISPR-Cas effector protein), a zinc finger nuclease, a transcription activator-like effector nuclease (TALEN) and/or an Argonaute protein)) and a deaminase or a nucleic acid construct encoding the same, under conditions whereby the sequence-specific nucleic acid binding protein, the reverse transcriptase and/or the deaminase are expressed and the sequence-specific nucleic acid binding protein binds to the target nucleic acid, and the reverse transcriptase and/or deaminase may be fused to either the sequence-specific nucleic acid binding protein or recruited to the sequence-specific nucleic acid binding protein (via, for example, a sequence-specific nu
  • modifying or “modification” in reference to a target nucleic acid includes editing (e.g., mutating), covalent modification, exchanging/substituting nucleic acids/nucleotide bases, deleting, cleaving, nicking, and/or altering transcriptional control of a target nucleic acid.
  • a modification may include one or more single base changes (SNPs) of any type.
  • introducing,” “introduce,” “introduced” in the context of a polynucleotide of interest means presenting a nucleotide sequence of interest (e.g., polynucleotide, RT template, a nucleic acid construct, and/or a guide nucleic acid) to a plant, plant part thereof, or cell thereof, in such a manner that the nucleotide sequence gains access to the interior of a cell.
  • a nucleotide sequence of interest e.g., polynucleotide, RT template, a nucleic acid construct, and/or a guide nucleic acid
  • a host cell or host organism e.g., a plant
  • a host cell or host organism may be transiently transformed with a polynucleotide/nucleic acid molecule of the invention.
  • Transient transformation in the context of a polynucleotide means that a polynucleotide is introduced into the cell and does not integrate into the genome of the cell.
  • stably introducing or “stably introduced” in the context of a polynucleotide introduced into a cell is intended that the introduced polynucleotide is stably incorporated into the genome of the cell, and thus the cell is stably transformed with the polynucleotide.
  • “Stable transformation” or “stably transformed” as used herein means that a nucleic acid molecule is introduced into a cell and integrates into the genome of the cell. As such, the integrated nucleic acid molecule is capable of being inherited by the progeny thereof, more particularly, by the progeny of multiple successive generations.
  • “Genome” as used herein includes the nuclear and the plastid genome, and therefore includes integration of the nucleic acid into, for example, the chloroplast or mitochondrial genome.
  • Stable transformation as used herein can also refer to a transgene that is maintained extrachromasomally, for example, as a minichromosome or a plasmid.
  • Transient transformation may be detected by, for example, an enzyme-linked immunosorbent assay (ELISA) or Western blot, which can detect the presence of a peptide or polypeptide encoded by one or more transgene introduced into an organism.
  • Stable transformation of a cell can be detected by, for example, a Southern blot hybridization assay of genomic DNA of the cell with nucleic acid sequences which specifically hybridize with a nucleotide sequence of a transgene introduced into an organism (e.g., a plant).
  • Stable transformation of a cell can be detected by, for example, a Northern blot hybridization assay of RNA of the cell with nucleic acid sequences which specifically hybridize with a nucleotide sequence of a transgene introduced into a host organism.
  • Stable transformation of a cell can also be detected by, e.g., a polymerase chain reaction (PCR) or other amplification reactions as are well known in the art, employing specific primer sequences that hybridize with target sequence(s) of a transgene, resulting in amplification of the transgene sequence, which can be detected according to standard methods. Transformation can also be detected by direct sequencing and/or hybridization protocols well known in the art.
  • PCR polymerase chain reaction
  • nucleotide sequences, polynucleotides, nucleic acid constructs, and/or expression cassettes of the invention may be expressed transiently and/or they can be stably incorporated into the genome of the host organism.
  • a nucleic acid construct of the invention e.g., one or more expression cassettes comprising polynucleotides for editing as described herein
  • a nucleic acid construct of the invention may be introduced into a plant cell by any method known to those of skill in the art.
  • transformation methods include transformation via bacterial-mediated nucleic acid delivery (e.g., via Agrobacteria), viral-mediated nucleic acid delivery, silicon carbide or nucleic acid whisker-mediated nucleic acid delivery, liposome mediated nucleic acid delivery, microinjection, microparticle bombardment, calcium-phosphate-mediated transformation, cyclodextrin-mediated transformation, electroporation, nanoparticle-mediated transformation, sonication, infiltration, PEG-mediated nucleic acid uptake, as well as any other electrical, chemical, physical (mechanical) and/or biological mechanism that results in the introduction of nucleic acid into the plant cell, including any combination thereof.
  • transformation of a cell may comprise nuclear transformation.
  • transformation of a cell may comprise plastid transformation (e.g., chloroplast transformation).
  • nucleic acids of the invention may be introduced into a cell via conventional breeding techniques.
  • one or more of the polynucleotides, expression cassettes and/or vectors may be introduced into a plant cell via Agrobacterium transformation.
  • a polynucleotide therefore can be introduced into a plant, plant part, plant cell in any number of ways that are well known in the art.
  • the methods of the invention do not depend on a particular method for introducing one or more nucleotide sequences into a plant, only that they gain access to the interior the cell.
  • they can be assembled as part of a single nucleic acid construct, or as separate nucleic acid constructs, and can be located on the same or different nucleic acid constructs.
  • the polynucleotide can be introduced into the cell of interest in a single transformation event, or in separate transformation events, or, alternatively, a polynucleotide can be incorporated into a plant as part of a breeding protocol.
  • the present invention provides methods and compositions for reducing the influence of genes that normally act to restrict meristem size to generate plants with larger meristems, to maintain the root meristem, to increase kernel row number (optionally without substantially decreasing ear length (e.g., without decreasing ear length more than 30% as compared to an ear of a plant not comprising the same CRN mutation)) and/or to improve one or more yield traits, as well as to improve disease resistance.
  • CLV1, CLV2, and CLV3 genes are part of several overlapping signaling pathways that utilize a similar mechanism to regulate meristem size.
  • Plants express many CLV3-like (CLE) peptides which are sensed by many leucine-rich repeat (LRR) domains, and the downstream signaling regulates a myriad of growth processes in the plant (Fletcher, J.C., Plants 7: 87 (2016)).
  • LRR leucine-rich repeat
  • CRN/CLV2 dimerization facilitates transit of both proteins from the endoplasmic reticulum (ER) to the plasma membrane (PM) where CLE-mediated signaling takes place.
  • ER endoplasmic reticulum
  • PM plasma membrane
  • CLV2 and CRN are separated as monomers, charged amino acids prevent PM transiting and the proteins are held in the ER.
  • CRN and CLV2 pair these charged amino acids are masked and export of both proteins from the ER to PM takes place, a phenomenon called charge masking (Bleckmann et ak Plant Phys.152: 166-176 (2010)).
  • the short extracellular domain (EC) of CRN is required for membrane transiting but is not required for dimerization with CLV2, suggesting the EC domain may perform a charge masking function. See, e.g., Fig. 4.
  • different amino acids can carry a negative, positive, or neutral charge.
  • the combined action of a sequence of amino acids determines the charge of a region of a particular protein.
  • the charge of the protein region that is involved in charge masking FEA2 can be changed and in-tum the transiting of the FEA2/CRN protein complex to the membrane can be modified.
  • the positive charge in CRN masks the negative charge of FEA2, which is required for transiting.
  • deletions of a CRN polypeptide may provide a reduction in positive charge, and thus, less charge masking of FEA2 and greater complex retention in the ER. Increased ER retention in this model would antagonize signaling mediated by the CRN/FEA2 complex resulting in increased meristem size, increased kernel row number and improved yield.
  • editing technology is used to target CRN genes in plants to generate plants with larger meristems, having increased kernel number (optionally without substantially reducing ear length, e.g., exhibits a decrease in ear length of no more than 30% as compared to an ear of a plant not comprising the same CRN mutation), increased yield, and improved disease resistance as well as to generate plants that maintain their root meristem.
  • Mutations that may be useful for production of plants exhibiting increased kernel number include, for example, substitutions, deletions and insertions.
  • a mutation generated by the editing technology can be a point mutation, an in-frame deletion, a mutation that generates a premature stop codon, a null mutation, a dominant negative mutation, a semi dominant mutation, hypomorphic mutation, or a weak loss-of-function mutation.
  • the invention provides a plant or plant part thereof, the plant or plant part comprising at least one non-natural mutation (e.g., 1, 2, 3, 4, 5, or more mutations) in an endogenous CORYNE (CRN) gene that encodes a CRN protein.
  • the at least one non-natural mutation results in a null mutation, a dominant negative mutation, a semi dominant mutation, a hypomorphic mutation, and/or a weak loss-of-function mutation.
  • a plant cell comprising an editing system comprising: (a) a CRISPR-Cas effector protein; and (b) a guide nucleic acid (gRNA, gDNA, crRNA, crDNA, sgRNA, sgDNA) comprising a spacer sequence with complementarity to an endogenous target gene encoding a CRN protein.
  • the editing system may be used to generate a mutation in the endogenous target gene encoding a CRN protein.
  • the mutation is a non-natural mutation.
  • a guide nucleic acid of an editing system may comprise the nucleotide sequence (a spacer sequence; guide) of any one of SEQ ID NOs:126-130.
  • the mutation in a CRN gene of the plant, plant part thereof or the plant cell may be any type of mutation, including a base substitution, a base deletion and/or a base insertion.
  • a non-natural mutation may comprise a base substitution to an A, a T, a G, or a C.
  • the at least one non-natural mutation may be a base substitution from a C to a T (C>T), a G to an A (G>A), an A to a G (A>G) or a T to a C (T>C).
  • a non-natural mutation may be a deletion of at least one base pair (e.g., 1 base pair to about 200 base pairs) or an insertion of at least one base pair (e.g., 1 base pair to about 50 base pairs).
  • a deletion may comprise 1 base pair to about 10 consecutive base pairs (e.g., 1, 2 bp to about 3, 4, 5, 6, 7, 8, 9, or 10 bp; e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 consecutive base pairs), 1 base pair to about 20 consecutive base pairs (e.g., 1, 2, 3, 4, 56, 7, 8,
  • a deletion may be a deletion that includes the entire EC region in a CRN gene (e.g., SEQ ID NO: 119 in maize).
  • a deletion is an in-frame deletion.
  • a deletion provides a premature stop codon and a truncated CRN gene and/or protein.
  • a non-natural mutation useful with this invention may results in at least one (e.g., one or more) amino acid substitution in the EC region of a CRN gene.
  • a non-natural mutation resulting in one or more substituted amino acid residues in a CRN gene may be located: (a) at position 23 to position 66 with reference amino acid position numbering of SEQ ID NO:118, (b) at position 29 to position 75 with reference amino acid position numbering of SEQ ID NOs:72-77; (c) at position 29 to position 74 with reference amino acid position numbering of SEQ ID NO:78; (d) at position 29 to position 76 with reference amino acid position numbering of SEQ ID NO:79; (e) at position 22 to position 65 with reference amino acid position numbering of SEQ ID NO:80; (1) at position 22 to position 68 with reference amino acid position numbering of SEQ ID NO:81; (g) at position 16 to position 65 with reference amino acid position numbering of SEQ ID NO:82; (h)
  • An endogenous CRN gene useful with this invention may (a) encode a sequence having at least 95% sequence identity to any one of the amino acid sequences of SEQ ID NO:72-118, optionally SEQ ID NO: 118; (b) comprise a region having a sequence with at least 90% sequence identity to the nucleotide sequence of SEQ ID NO:122-125; and/or (c) encode a sequence having at least 90% sequence identity to any one of the amino acid sequences of SEQ ID NOs: 119-121.
  • a mutated CRN gene comprises at least about 90% sequence identity to any one of the nucleic acid sequences of SEQ ID NOs:132-145.
  • a plant comprising at least one mutation (e.g., one or more mutations) in an endogenous CRN gene exhibits increased maintenance of meristems and/or increased kernel number as compared to a plant without the at least one non-natural mutation.
  • the plant comprising at least one mutation in an endogenous CRN gene is a com plant which exhibits increased kernel number. In some embodiments, the plant comprising at least one mutation in an endogenous CRN gene is a com plant which exhibits increased yield and/or increased disease resistance. In some embodiments, a plant (e.g., a com plant) may be regenerated from a plant part and/or plant cell of the invention, wherein the regenerated plant (e.g., regenerated com plant) comprises the mutation in the endogenous CRN gene and a phenotype of increased kernel number as compared to a plant (e.g., a com plant) not comprising the mutation.
  • a plant e.g., a com plant
  • a com plant cell comprising at least one non-natural mutation within a CRN gene, wherein the mutation is a substitution, insertion or a deletion that is introduced using an editing system that comprises a nucleic acid binding domain that binds to a target site in the CRN gene.
  • the substitution, insertion or a deletion results in a null allele, a dominant negative allele, a semi dominant allele, hypomorphic mutation, or a weak loss-of-function allele.
  • a deletion results in an in-frame deletion allele.
  • the target site is within a region of the CRN gene, the region comprising a sequence having at least 90% sequence identity (e.g., about 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 99.5, 99.6, 99.7, 99.8, 99.9, or 100% sequence identity) to the nucleotide sequence of any one of SEQ ID NOs:123-125 and/or encoding a sequence having at least 95% sequence identity (e.g., about 95, 96, 97, 98, 99, 99.5, 99.5, 99.6, 99.7, 99.8, 99.9, or 100% sequence identity) to any one of the amino acid sequences of SEQ ID NO: 119-121.
  • a sequence having at least 90% sequence identity e.g., about 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 99.5, 99.6, 99.7, 99.8, 99.9, or 100% sequence identity
  • a method of producing/breeding a transgene-free edited com plant comprising: crossing a com plant of the present invention (e.g., a com plant comprising a mutation in a CRN gene and having increased kernel number) with a transgene free com plant, thereby introducing the at least one non-natural mutation (e.g., one or more non-natural mutations) into the com plant that is transgene-free; and selecting a progeny com plant that comprises the at least one non-natural mutation and is transgene-free, thereby producing a transgene free edited com plant.
  • a com plant of the present invention e.g., a com plant comprising a mutation in a CRN gene and having increased kernel number
  • the at least one non-natural mutation e.g., one or more non-natural mutations
  • Also provided herein is a method of providing a plurality of com plants having increased kernel number, the method comprising planting two or more com plants of the invention (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10 or more com plants comprising a mutation in a CRN polypeptide and having increased kernel number) in a growing area (e.g., a field (e.g., a cultivated field, an agricultural field), a growth chamber, a greenhouse, a recreational area, a lawn, and/or a roadside and the like), thereby providing a plurality of com plants having increased kernel number as compared to a plurality of control com plants not comprising the mutation.
  • a growing area e.g., a field (e.g., a cultivated field, an agricultural field), a growth chamber, a greenhouse, a recreational area, a lawn, and/or a roadside and the like
  • the plurality of plants may also exhibit larger meristems, increased yield, increased disease resistance and maintenance of their root meristems.
  • the invention further provides a method of generating variation in a region of a CRN protein, comprising: introducing an editing system into a com plant cell, wherein the editing system is targeted to a region of a CRN gene that encodes the region of the CRN protein, wherein the region comprises a sequence having at least 70% sequence identity to any one of the amino acid sequences of SEQ ID NO:119-121 or the region is encoded by a sequence having at least 70% identity to the nucleotide sequence of SEQ ID NOs:123-125; and contacting the region of the CRN gene with the editing system, thereby introducing into the plant cell a mutation into the region of the CRN protein; and generating variation in the region of the CRN protein.
  • a method for editing a specific site in the genome of a com plant cell comprising: cleaving, in a site specific manner, a target site within an endogenous CRN gene in the plant cell, the endogenous CRN gene comprising a sequence having at least 70% sequence identity to the nucleotide sequence of SEQ ID NO: 122, or encoding a sequence having at least 70% sequence identity to the amino acid sequence of SEQ ID NO:118, thereby generating an edit in the endogenous CRN gene of the plant cell and producing a plant cell comprising the edit in the endogenous CRN gene.
  • the endogenous CRN gene encodes a CRN protein and the edit results in variation of amino acids in the coding region of the CRN protein.
  • the edit results in a non-naturally occurring mutation, including but not limited to a deletion, substitution, or insertion, wherein the edit may result in a null mutation, a dominant negative mutation, a semi-dominant mutation, a hypomorphic mutation, or a weak loss-of-function mutation.
  • the non- naturally occurring mutation is a deletion, optionally wherein the deletion is in the EC domain of a CRN gene (See, Fig. 3 - representation of various deletions within the EC domain).
  • the entire EC domain of the CRN gene may be deleted.
  • the deletion may result in an edited CRN nucleic acid having at least 90% sequence identity to any one of SEQ ID NOs: 132-145.
  • a deletion results in an in-frame deletion.
  • a deletion results in the generation of a premature stop codon.
  • anon-natural mutation produces variability in a region (e.g., EC domain) of a CRN polypeptide (e.g., amino acid residues located at position 23 to position 66 with reference amino acid position numbering of SEQ ID NO:118 and/or in the EC region of any one of the amino acid sequences of SEQ ID NOs:72-117 as described herein).
  • a region e.g., EC domain
  • CRN polypeptide e.g., amino acid residues located at position 23 to position 66 with reference amino acid position numbering of SEQ ID NO:118 and/or in the EC region of any one of the amino acid sequences of SEQ ID NOs:72-117 as described herein.
  • a method of editing may further comprise regenerating a com plant from the com plant cell comprising the edit in the endogenous CRN gene, thereby producing a com plant comprising the edit in its endogenous CRN gene and having a phenotype of increased kernel number when compared to a control com plant (e.g., an isogenic com plant) that does not comprise the edit.
  • a control com plant e.g., an isogenic com plant
  • a method for making a com plant comprising: (a) contacting a population of com plant cells comprising a wild-type endogenous CRN gene with a nuclease linked to a nucleic acid binding domain (e.g., DNA binding domain; e.g., editing system) that binds to a sequence having at least 70% sequence identity to the nucleotide sequence of SEQ ID NO:122, to a sequence having at least 70% identity to the nucleotide sequence of any one of SEQ ID NOs: 123-125, to a sequence encoding an amino acid sequence having at least 70% sequence identity to SEQ ID NO: 118; or to a sequence encoding an amino acid sequence having at least 70% sequence identity to any one of SEQ ID NOs:119-121; (b) selecting a com plant cell from the population in which at least one endogenous CRN gene has been mutated; and (c) growing the selected plant cell into a com plant.
  • a nucleic acid binding domain e
  • a method increasing kernel number in a com plant comprising (a) contacting a com plant cell comprising an endogenous CRN gene with a nuclease targeting the endogenous CRN gene, wherein the nuclease is linked to a nucleic acid binding domain (e.g., editing system) that binds to a target site in the endogenous CRN gene, wherein the endogenous CRN gene: (i) encodes a sequence having at least 70% sequence identity to the amino acid sequence of SEQ ID NO:118; (ii) comprises a sequence having at least 70% sequence identity to the nucleotide sequence of SEQ ID NO: 122; (iii) comprises a region having a sequence with at least 70% sequence identity to the nucleotide sequence of any one of SEQ ID NOs:123-125; and/or (iv) comprises a region encoding a sequence having at least 70% sequence identity to the amino acid sequence of SEQ ID NOs:119-121 to produce a
  • a method for producing a com plant or part thereof comprising at least one cell (e.g., one or more cells) having a mutated endogenous CRN gene comprising contacting a target site in an endogenous CRN gene in the com plant or plant part with a nuclease comprising a cleavage domain and a nucleic acid binding domain, wherein the nucleic acid binding domain binds to a target site in the endogenous CRN gene, wherein the endogenous CRN gene (a) encodes a sequence having at least 70% sequence identity to the amino acid sequence of SEQ ID NO:118; (b) comprises a sequence having at least 70% sequence identity to the nucleotide sequence of SEQ ID NO:122; (c) comprises a region having a sequence with at least 70% sequence identity to the nucleotide sequence of any one of SEQ ID NOs:123-125; and/or (d) comprises a region encoding a sequence having at least 70% sequence identity to the
  • Also provided herein is a method for producing a com plant or part thereof comprising a mutated endogenous CRN gene and exhibiting increased kernel number, the method comprising contacting a target site in an endogenous CRN gene in the com plant or plant part with a nuclease comprising a cleavage domain and a nucleic acid binding domain, wherein the nucleic acid binding domain binds to a target site in the endogenous CRN gene, wherein the endogenous CRN gene: (a) encodes a sequence having at least 70% sequence identity to the amino acid sequence of SEQ ID NO:118; (b) comprises a sequence having at least 70% sequence identity to the nucleotide sequence of SEQ ID NO: 122; (c) comprises a region having a sequence with at least 70% sequence identity to the nucleotide sequence of any one of SEQ ID NOs:123-125; and/or (d) comprises a region encoding a sequence having at least 70% sequence identity to the amino acid sequence of SEQ ID
  • a com plant or part thereof comprising at least one cell (e.g., one or more cells) having a mutation in the endogenous CRN gene as described herein comprises a sequence having at least 90% identity to any one of the sequences of SEQ ID NOs: 132-145.
  • a nuclease may cleave an endogenous CRN gene, thereby introducing the mutation into the endogenous CRN gene.
  • a nuclease useful with the invention may be any nuclease that can be utilized to edit/modify a target nucleic acid.
  • Such nucleases include, but are not limited to a zinc finger nuclease, transcription activator-like effector nucleases (TALEN), endonuclease (e.g., Fokl) and/or a CRISPR-Cas effector protein.
  • nucleic acid binding domain useful with the invention may be any nucleic acid binding domain (e.g., DNA binding domain) that can be utilized to edit/modify a target nucleic acid.
  • nucleic acid binding domains include, but are not limited to, a zinc finger, transcription activator-like DNA binding domain (TAL), an argonaute and/or a CRISPR-Cas effector DNA binding domain.
  • a method of editing an endogenous CRN gene in a com plant or plant part comprising contacting a target site in an CRN gene in the com plant or plant part with a cytosine base editing system comprising a cytosine deaminase and a nucleic acid binding domain that binds to a target site in the CRN gene, the CRN gene (a) encoding a sequence having at least 70% sequence identity to the amino acid sequence of SEQ ID NO:118; (b) comprising a sequence having at least 70% sequence identity to the nucleotide sequence of SEQ ID NO:122; (c) comprising a region having a sequence with at least 70% sequence identity to the nucleotide sequence of any one of SEQ ID NOs: 123-125; and/or (d) comprising a region encoding a sequence having at least 70% sequence identity to the amino acid sequence of SEQ ID NOs: 119-121, thereby editing the endogenous CRN gene in the
  • a method of editing an endogenous CRN gene in a com plant or plant part comprising contacting a target site in an CRN gene in the com plant or plant part with an adenosine base editing system comprising an adenosine deaminase and a nucleic acid binding domain that binds to a target site in the CRN gene, the CRN gene (a) encoding a sequence having at least 70% sequence identity to the amino acid sequence of SEQ ID NO:118; (b) comprising a sequence having at least 70% sequence identity to the nucleotide sequence of SEQ ID NO:122; (c) comprising a region having a sequence with at least 70% sequence identity to the nucleotide sequence of any one of SEQ ID NOs: 123-125; and/or (d) comprising a region encoding a sequence having at least 70% sequence identity to the amino acid sequence of SEQ ID NOs: 119-121, thereby editing the endogenous CRN gene
  • a mutation in an edited endogenous CRN gene as described herein produces a sequence having at least 90% identity to any one of the sequences of SEQ ID
  • a method of detecting a mutant CRN gene (a mutation in an endogenous CRN gene) is provide, the method comprising detecting in the genome of a plant a deletion in a nucleic acid encoding the amino acid sequence of SEQ ID NO: 118, wherein the amino acid sequence of SEQ ID NO: 118 comprises a mutation in one or more amino acid residue(s) located from position 23 to position 66 with reference amino acid position numbering of SEQ ID NO:118.
  • the mutation is the result of a nucleotide substitution of OT, G>A, A>G or T>C.
  • the present invention provides a method of detecting a mutation in an endogenous CRN gene, comprising detecting in the genome of a plant a mutated CRN gene.
  • the mutated CRN gene comprises a sequence having at least 90% sequence identity to the nucleotide sequence of any one of SEQ ID NOs: 132-145.
  • the present invention provides a method of producing a plant comprising a mutation in an endogenous CRN gene and at least one polynucleotide of interest (e.g., one or more polynucleotides of interest), the method comprising crossing a plant of the invention comprising at least one mutation in an endogenous CRN gene (a first plant) with a second plant that comprises the at least one polynucleotide of interest to produce progeny plants; and selecting progeny plants comprising at least one mutation in the CRN gene and the at least one polynucleotide of interest, thereby producing the plant comprising a mutation in an endogenous CRN gene and at least one polynucleotide of interest.
  • a polynucleotide of interest e.g., one or more polynucleotides of interest
  • the present invention further provides a method of producing a plant comprising a mutation in an endogenous CRN gene and at least one polynucleotide of interest, the method comprising introducing at least one polynucleotide of interest into a plant of the present invention comprising at least one mutation in a CRN gene, thereby producing a plant comprising at least one mutation in a CRN gene and at least one polynucleotide of interest.
  • the plant is a com plant.
  • the present invention provides a method of producing a plant comprising a mutation in an endogenous CRN gene and at least one polynucleotide of interest, the method comprising introducing at least one polynucleotide of interest into a plant of the invention comprising at least one mutation in an endogenous CRN gene, thereby producing a plant comprising at least one mutation in a CRN gene and at least one polynucleotide of interest.
  • the plant is a com plant.
  • a polynucleotide of interest may be any polynucleotide that can confer a desirable phenotype or otherwise modify the phenotype or genotype of a plant.
  • a polynucleotide of interest may be polynucleotide that confers herbicide tolerance, insect resistance, disease resistance, increased yield, increased nutrient use efficiency or abiotic stress resistance.
  • a CORYNE (CRN) gene useful with this invention includes any CRN gene in which a mutation as described herein can confer increased kernel number in a plant or part thereof comprising the mutation (e.g., SEQ ID NOs:72-118).
  • a CRN polypeptide comprises an amino acid sequence having at least 70% identity (e.g., about 70, 71, 72, 73, 74, 75, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 99.5,
  • the CRN polypeptide comprises an EC domain comprising the amino acid sequence of, for example, SEQ ID NO:119, SEQ ID NO:120, or SEQ ID NO:121) within the CRN polypeptide).
  • a CRN polypeptide comprises an amino acid sequence having at least 70% identity (e.g., about 70, 71, 72, 73, 74, 75, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 99.5, 100% sequence identity) to SEQ ID NO:72-118.
  • a CNR gene may comprise a sequence having at least about 70% sequence identity (e.g., about 70, 71, 72, 73, 74, 75, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 99.5, 100% sequence identity) to the nucleotide sequence of SEQ ID NO: 122, or the CRN gene comprises within it a sequence having at least 70% identity to any one of the nucleotide sequences of any one of SEQ ID NOs: 123-125
  • the at least one non-natural mutation (e.g., one or more non natural mutations) in an endogenous CRN gene in a com plant may be a substitution, a deletion and/or an insertion.
  • the at least one non-natural mutation in an endogenous CRN gene in a com plant may be a substitution, a deletion and/or an insertion that results in a null mutation, a dominant negative mutation, a semi-dominant mutation, hypomorphic mutation, or a weak loss-of-function mutation and a plant having the phenotype of increased kernel number as compared to a control com plant (e.g., isogenic plant) not comprising the edit/mutation.
  • the mutation may be a substitution, a deletion and/or an insertion of one or more amino acid residues (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more amino acids of the CRN polypeptide) or the mutation may be a substitution, a deletion and/or an insertion of at least 1 nucleotide to about 160 consecutive nucleotides (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50 consecutive nucleotides, or any range or value therein) (e.g., a base substitution, deletion and/or insertion) from the gene encoding the CRN polypeptide.
  • the non-natural mutation may be a deletion.
  • the mutation may be a deletion of at least about 18 consecutive base pairs to about 160 consecutive base
  • a deletion results in an in-frame deletion allele.
  • the at least one non-natural mutation may be a base substitution to an A, a T, a G, or a C.
  • the at least one non-natural mutation may be a base substitution to from a C to a T (C>T), a G to an A (G>A), an A to a G (A>G) or a T to a C (T>C).
  • the non-natural mutation is in the EC region of a CRN.
  • a mutation in an endogenous CRN gene may be made following cleavage by an editing system that comprises a nuclease and a nucleic acid binding domain that binds to a target site within a target nucleic acid (e.g., a CRN gene), the target nucleic acid (e.g., CRN gene) (a) encoding a sequence having at least 70% sequence identity to the amino acid sequence of SEQ ID NO:118; (b) comprising a sequence having at least 70% sequence identity to the nucleotide sequence of SEQ ID NO: 122; (c) comprising a region having a sequence with at least 70% sequence identity to the nucleotide sequence of any one of SEQ ID NOs:123-125; and/or (d) comprising a region encoding a sequence having at least 70% sequence identity to the amino acid sequence of SEQ ID NOs:119-121.
  • an editing system that comprises a nuclease and a nucleic acid binding domain that binds to a
  • the nuclease cleaves the endogenous CRN gene, and a mutation is introduced into the endogenous CRN gene. In some embodiments, the cleavage results in a mutation in an endogenous CRN gene comprising a sequence having at least 90% identity to any one of the sequences of SEQ ID NOs: 132-145.
  • guide nucleic acids e.g., gRNA, gDNA, crRNA, crDNA
  • the endogenous CRN gene comprises: a sequence having at least 70% sequence identity to the nucleotide sequence of SEQ ID NO:122 or any one of the nucleotide sequences of any one of SEQ ID NOs:123-125; encodes a sequence having at least 70% sequence identity to the amino acid sequence of SEQ ID NO:118, or any one of the amino acid sequences of SEQ ID NO: 119-121.
  • a guide nucleic acid comprises a spacer having the nucleotide sequence of any one of SEQ ID NOs: 126-130.
  • a guide nucleic acid that binds to a target nucleic acid in a CORYNE (CRN) gene in a com plant, the CRN gene having the gene identification number (gene ID) of Zm00001d042268.
  • CRN CORYNE
  • markers of the present invention are described herein with respect to the positions of marker loci in the B73 com genome, version 4, "B73 RefGen_v4" (assembly aka B73 RefGen_v4, AGPv4) at the MaizeGDB internet resource (maizegdb.org/assembly).
  • a system comprising a guide nucleic acid comprising a spacer having the nucleotide sequence of any one of SEQ ID NOs: 126-130 and a CRISPR- Cas effector protein that associates with the guide nucleic acid.
  • the system may further comprise a tracr nucleic acid that associates with the guide nucleic acid and a CRISPR-Cas effector protein, optionally wherein the tracr nucleic acid and the guide nucleic acid are covalently linked.
  • the invention further provides a gene editing system comprising a CRISPR-Cas effector protein in association with a guide nucleic acid and the guide nucleic acid comprises a spacer sequence that binds to a CRN gene, the CRN gene (a) encoding a sequence having at least 70% sequence identity to the amino acid sequence of SEQ ID NO:118; (b) comprising a sequence having at least 70% sequence identity to the nucleotide sequence of SEQ ID NO: 122; (c) comprising a region having a sequence with at least 70% sequence identity to the nucleotide sequence of any one of SEQ ID NOs: 123-125; and/or (d) comprising a region encoding a sequence having at least 70% sequence identity to the amino acid sequence of SEQ ID NOs: 119-121.
  • a spacer sequence of the guide nucleic acid may comprise the nucleotide sequence of any one of SEQ ID NOs: 126-130.
  • the gene editing system may further comprise a tracr nucleic acid that associates with the guide nucleic acid and a CRISPR-Cas effector protein, optionally wherein the tracr nucleic acid and the guide nucleic acid are covalently linked.
  • a CRISPR-Cas effector protein in association with a guide nucleic acid refers to the complex that is formed between a CRISPR- Cas effector protein and a guide nucleic acid in order to direct the CRISPR-Cas effector protein to a target site in a gene.
  • the present invention further provides a complex comprising a CRISPR-Cas effector protein comprising a cleavage domain and a guide nucleic acid, wherein the guide nucleic acid binds to a target site in an endogenous CRN gene, wherein the endogenous CRN gene, wherein the endogenous CRN gene, wherein the endogenous CRN gene: (a) encodes a sequence having at least 70% sequence identity to the amino acid sequence of SEQ ID NO:118; (b) comprises a sequence having at least 70% sequence identity to the nucleotide sequence of SEQ ID NO:122; (c) comprises a region having a sequence with at least 70% sequence identity to the nucleotide sequence of any one of SEQ ID NOs:123-125; and/or (d) comprises a region encoding a sequence having at least 70% sequence identity to the amino acid sequence of SEQ ID NOs: 119-121, wherein the cleavage domain cleaves a target strand in the CRN gene.
  • the cleavage domain cleaves a target strand in the CRN gene such that it results in a mutation in an endogenous CRN gene, the mutated CRN gene comprising a sequence having at least 90% identity to any one of the sequences of SEQ ID NOs:132-145.
  • expression cassettes comprise (a) a polynucleotide encoding CRISPR-Cas effector protein comprising a cleavage domain and (b) a guide nucleic acid that binds to a target site in an endogenous CRN gene, wherein the guide nucleic acid comprises a spacer sequence that is complementary to and binds to (i) a portion of a nucleic acid encoding an amino acid sequence having at least 70% sequence identity the amino acid sequence of SEQ ID NO:118; (ii) a portion of a sequence having at least 70% sequence identity to the nucleotide sequence of SEQ ID NO: 122; (iii) a portion of a sequence having at least 70% sequence identity to the nucleotide sequence of any one of SEQ ID NOs:123-125; and/or (iv) a portion of sequence having at least 70% sequence identity to a sequence encoding the amino acid sequence of SEQ ID NO:119-121.
  • nucleic acids encoding a mutated CRN gene that when present in a com plant or plant part results in the com plant comprising a phenotype of increased kernel number as compared to a com plant or plant part not comprising the CRN mutation.
  • a mutated CRN gene may comprise a sequence having at least 90% sequence identity to the nucleotide sequence of any one of SEQ ID NOs: 132-145.
  • Nucleic acid constructs of the invention e.g., a construct comprising a sequence specific nucleic acid binding domain, a CRISPR-Cas effector domain, a deaminase domain, reverse transcriptase (RT), RT template and/or a guide nucleic acid, etc.
  • expression cassettes/vectors comprising the same may be used as an editing system of this invention for modifying target nucleic acids (e.g., endogenous CRN genes) and/or their expression.
  • Any com plant comprising an endogenous CRN gene that is capable of conferring increased kernel number when modified as described herein may be modified (e.g., mutated, e.g., base edited, cleaved, nicked, etc.) as described herein (e.g., using the polypeptides, polynucleotides, RNPs, nucleic acid constructs, expression cassettes, and/or vectors of the invention) to increase kernel number in the com plant.
  • a plant exhibiting increased kernel number may have an increase in kernel number by about 5% to about 100% (e.g., about 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16,
  • plants exhibiting increased kernel row number as described herein produce ears that are also not substantially decreased in length.
  • an ear of a plant comprising a mutation as described herein that is "not substantially decreased in length” has a length that is reduced by less than 30% (e.g., reduced by 0% or reduced by about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10,
  • a com plant or plant part thereof comprising at least one non-natural mutation in at least one endogenous CORYNE (CRN) gene having the gene identification number (gene ID) of Zm00001d042268.
  • CRN CORYNE
  • plant part includes reproductive tissues (e.g., petals, sepals, stamens, pistils, receptacles, anthers, pollen, flowers, fruits, flower bud, ovules, seeds, and embryos); vegetative tissues (e.g., petioles, stems, roots, root hairs, root tips, pith, coleoptiles, stalks, shoots, branches, bark, apical meristem, axillary bud, cotyledon, hypocotyls, and leaves); vascular tissues (e.g., phloem and xylem); specialized cells such as epidermal cells, parenchyma cells, chollenchyma cells, schlerenchyma cells, stomates, guard cells, cuticle, mesophyll cells; callus tissue; and cuttings.
  • reproductive tissues e.g., petals, sepals, stamens, pistils, receptacles, anthers,
  • plant part also includes plant cells, including plant cells that are intact in plants and/or parts of plants, plant protoplasts, plant tissues, plant organs, plant cell tissue cultures, plant calli, plant clumps, and the like.
  • shoot refers to the above ground parts including the leaves and stems.
  • tissue culture encompasses cultures of tissue, cells, protoplasts and callus.
  • plant cell refers to a structural and physiological unit of the plant, which typically comprise a cell wall but also includes protoplasts.
  • a plant cell of the present invention can be in the form of an isolated single cell or can be a cultured cell or can be a part of a higher-organized unit such as, for example, a plant tissue (including callus) or a plant organ.
  • a "protoplast” is an isolated plant cell without a cell wall or with only parts of the cell wall.
  • a transgenic cell comprising a nucleic acid molecule and/or nucleotide sequence of the invention is a cell of any plant or plant part including, but not limited to, a root cell, a leaf cell, a tissue culture cell, a seed cell, a flower cell, a fruit cell, a pollen cell, and the like.
  • the plant part can be a plant germplasm.
  • a plant cell can be non-propagating plant cell that does not regenerate into a plant.
  • Plant cell culture means cultures of plant units such as, for example, protoplasts, cell culture cells, cells in plant tissues, pollen, pollen tubes, ovules, embryo sacs, zygotes and embryos at various stages of development.
  • a "plant organ” is a distinct and visibly structured and differentiated part of a plant such as a root, stem, leaf, flower bud, or embryo.
  • Plant tissue as used herein means a group of plant cells organized into a structural and functional unit. Any tissue of a plant in planta or in culture is included. This term includes, but is not limited to, whole plants, plant organs, plant seeds, tissue culture and any groups of plant cells organized into structural and/or functional units. The use of this term in conjunction with, or in the absence of, any specific type of plant tissue as listed above or otherwise embraced by this definition is not intended to be exclusive of any other type of plant tissue.
  • transgenic tissue culture or transgenic plant cell culture wherein the transgenic tissue or cell culture comprises a nucleic acid molecule/nucleotide sequence of the invention.
  • transgenes may be eliminated from a plant developed from the transgenic tissue or cell by breeding of the transgenic plant with a non-transgenic plant and selecting among the progeny for the plants comprising the desired gene edit and not the transgenes used in producing the edit.
  • An editing system useful with this invention can be any site-specific (sequence-specific) genome editing system now known or later developed, which system can introduce mutations in target specific manner.
  • an editing system e.g., site- or sequence-specific editing system
  • CRISPR-Cas editing system e.g., a meganuclease editing system
  • ZFN zinc finger nuclease
  • TALEN transcription activator-like effector nucle
  • an editing system e.g., site- or sequence-specific editing system
  • an editing system can comprise one or more sequence-specific nucleic acid binding domains (e.g., DNA binding domains) that can be from, for example, a polynucleotide-guided endonuclease, a CRISPR-Cas endonuclease (e.g., CRISPR-Cas effector protein), a zinc finger nuclease, a transcription activator-like effector nuclease (TALEN) and/or an Argonaute protein.
  • sequence-specific nucleic acid binding domains e.g., DNA binding domains
  • an editing system can comprise one or more cleavage domains (e.g., nucleases) including, but not limited to, an endonuclease (e.g., Fokl), a polynucleotide-guided endonuclease, a CRISPR-Cas endonuclease (e.g., CRISPR-Cas effector protein), a zinc finger nuclease, and/or a transcription activator-like effector nuclease (TALEN).
  • nucleases including, but not limited to, an endonuclease (e.g., Fokl), a polynucleotide-guided endonuclease, a CRISPR-Cas endonuclease (e.g., CRISPR-Cas effector protein), a zinc finger nuclease, and/or a transcription activator-like effector nuclease (TALEN).
  • an editing system can comprise one or more polypeptides that include, but are not limited to, a deaminase (e.g., a cytosine deaminase, an adenine deaminase), a reverse transcriptase, a Dna2 polypeptide, and/or a 5' flap endonuclease (FEN).
  • a deaminase e.g., a cytosine deaminase, an adenine deaminase
  • a reverse transcriptase e.g., a reverse transcriptase
  • Dna2 polypeptide e.g., a 5' flap endonuclease (FEN).
  • FEN 5' flap endonuclease
  • an editing system can comprise one or more polynucleotides, including, but is not limited to, a CRISPR array (CRISPR guide) nucleic acid, extended guide nucleic acid,
  • a method of modifying or editing an CRN gene may comprise contacting a target nucleic acid (e.g., a nucleic acid encoding an CRN protein) with a base editing fusion protein (e.g., a sequence specific DNA binding protein (e.g., a CRISPR-Cas effector protein or domain) fused to a deaminase domain (e.g., an adenine deaminase and/or a cytosine deaminase) and a guide nucleic acid, wherein the guide nucleic acid is capable of guiding/targeting the base editing fusion protein to the target nucleic acid, thereby editing a locus within the target nucleic acid.
  • a target nucleic acid e.g., a nucleic acid encoding an CRN protein
  • a base editing fusion protein e.g., a sequence specific DNA binding protein (e.g., a CRISPR-Cas effector protein or domain
  • a base editing fusion protein and guide nucleic acid may be comprised in one or more expression cassettes.
  • the target nucleic acid may be contacted with a base editing fusion protein and an expression cassette comprising a guide nucleic acid.
  • the sequence- specific nucleic acid binding fusion proteins and guides may be provided as ribonucleoproteins (RNPs).
  • a cell may be contacted with more than one base-editing fusion protein and/or one or more guide nucleic acids that may target one or more target nucleic acids in the cell.
  • a method of modifying or editing an CRN gene may comprise contacting a target nucleic acid (e.g., a nucleic acid encoding a CRN) with a sequence-specific nucleic acid binding fusion protein (e.g., a sequence-specific DNA binding protein (e.g., a CRISPR-Cas effector protein or domain)) fused to a peptide tag, a deaminase fusion protein comprising a deaminase domain (e.g., an adenine deaminase and/or a cytosine deaminase) fused to an affinity polypeptide that is capable of binding to the peptide tag, and a guide nucleic acid, wherein the guide nucleic acid is capable of guiding/targeting the sequence-specific nucleic acid binding fusion protein to the target nucleic acid and the sequence-specific nucleic acid binding fusion protein is capable of recruiting the deaminase fusion protein to
  • the sequence-specific nucleic acid binding fusion protein may be fused to the affinity polypeptide that binds the peptide tag and the deaminase may be fuse to the peptide tag, thereby recruiting the deaminase to the sequence-specific nucleic acid binding fusion protein and to the target nucleic acid.
  • the sequence-specific binding fusion protein, deaminase fusion protein, and guide nucleic acid may be comprised in one or more expression cassettes.
  • the target nucleic acid may be contacted with a sequence-specific binding fusion protein, deaminase fusion protein, and an expression cassette comprising a guide nucleic acid.
  • the sequence- specific nucleic acid binding fusion proteins, deaminase fusion proteins and guides may be provided as ribonucleoproteins (RNPs).
  • methods such as prime editing may be used to generate a mutation in an endogenous CRN gene.
  • prime editing RNA-dependent DNA polymerase (reverse transcriptase, RT) and reverse transcriptase templates (RT template) are used in combination with sequence specific nucleic acid binding domains that confer the ability to recognize and bind the target in a sequence-specific manner, and which can also cause a nick of the PAM-containing strand within the target.
  • the nucleic acid binding domain may be a CRISPR-Cas effector protein and in this case, the CRISPR array or guide RNA may be an extended guide that comprises an extended portion comprising a primer binding site (PSB) and the edit to be incorporated into the genome (the template).
  • PSB primer binding site
  • prime editing can take advantageous of the various methods of recruiting proteins for use in the editing to the target site, such methods including both non-covalent and covalent interactions between the proteins and nucleic acids used in the selected process of genome editing.
  • a "CRISPR-Cas effector protein” is a protein or polypeptide or domain thereof that cleaves or cuts a nucleic acid, binds a nucleic acid (e.g., a target nucleic acid and/or a guide nucleic acid), and/or that identifies, recognizes, or binds a guide nucleic acid as defined herein.
  • a CRISPR-Cas effector protein may be an enzyme (e.g., a nuclease, endonuclease, nickase, etc.) or portion thereof and/or may function as an enzyme.
  • a CRISPR-Cas effector protein refers to a CRISPR-Cas nuclease polypeptide or domain thereof that comprises nuclease activity or in which the nuclease activity has been reduced or eliminated, and/or comprises nickase activity or in which the nickase has been reduced or eliminated, and/or comprises single stranded DNA cleavage activity (ss DNAse activity) or in which the ss DNAse activity has been reduced or eliminated, and/or comprises self-processing RNAse activity or in which the self-processing RNAse activity has been reduced or eliminated.
  • a CRISPR-Cas effector protein may bind to a target nucleic acid.
  • a sequence-specific nucleic acid binding domain may be a CRISPR-Cas effector protein.
  • a CRISPR-Cas effector protein may be from a Type I CRISPR-Cas system, a Type II CRISPR-Cas system, a Type III CRISPR-Cas system, a Type IV CRISPR-Cas system, Type V CRISPR-Cas system, or a Type VI CRISPR- Cas system.
  • a CRISPR-Cas effector protein of the invention may be from a Type II CRISPR-Cas system or a Type V CRISPR-Cas system.
  • a CRISPR-Cas effector protein may be Type II CRISPR-Cas effector protein, for example, a Cas9 effector protein.
  • a CRISPR-Cas effector protein may be Type V CRISPR-Cas effector protein, for example, a Casl2 effector protein.
  • a CRISPR-Cas effector protein may include, but is not limited to, a Cas9, C2cl, C2c3, Casl2a (also referred to as Cpfl), Casl2b, Casl2c, Casl2d, Casl2e, Casl3a, Casl3b, Casl3c, Casl3d, Casl, CaslB, Cas2, Cas3, Cas3', Cas3", Cas4, Cas5, Cas6, Cas7, Cas8, Cas9 (also known as Csnl and Csxl2), CaslO, Csyl, Csy2, Csy3, Csel, Cse2, Cscl, Csc2, Csa5, Csn2, Csm2, Csm3, Csm4, Csm5, Csm6, Cmrl, Cmr3, Cmr4, Cmr5, Cmr6, Csbl, Csb2,
  • a CRISPR-Cas effector protein useful with the invention may comprise a mutation in its nuclease active site (e.g., RuvC, HNH, e.g., RuvC site of a Casl2a nuclease domain; e.g., RuvC site and/or HNH site of a Cas9 nuclease domain).
  • a CRISPR-Cas effector protein having a mutation in its nuclease active site, and therefore, no longer comprising nuclease activity is commonly referred to as "dead,” e.g., dCas.
  • a CRISPR-Cas effector protein domain or polypeptide having a mutation in its nuclease active site may have impaired activity or reduced activity as compared to the same CRISPR-Cas effector protein without the mutation, e.g., a nickase, e.g, Cas9 nickase, Casl2a nickase.
  • a CRISPR Cas9 effector protein or CRISPR Cas9 effector domain useful with this invention may be any known or later identified Cas9 nuclease.
  • a CRISPR Cas9 polypeptide can be a Cas9 polypeptide from, for example, Streptococcus spp. (e.g., S. pyogenes, S. thermophilus), Lactobacillus spp., Bifidobacterium spp., Kandleria spp., Leuconostoc spp., Oenococcus spp., Pediococcus spp., Weissella spp., and/or Olsenella spp.
  • Streptococcus spp. e.g., S. pyogenes, S. thermophilus
  • Lactobacillus spp. e.g., S. pyogenes, S. thermophilus
  • Example Cas9 sequences include, but are not limited to, the amino acid sequences of SEQ ID NO:59 and SEQ ID NO:60 or the nucleotide sequences of SEQ ID NOs:61-71.
  • the CRISPR-Cas effector protein may be a Cas9 polypeptide derived from Streptococcus pyogenes and recognizes the PAM sequence motif NGG, NAG, NGA (Mali et al, Science 2013; 339(6121): 823-826).
  • the CRISPR-Cas effector protein may be a Cas9 protein derived from S.
  • N can be any nucleotide residue, e.g., any of A, G, C or T.
  • the CRISPR-Cas effector protein may be a Casl3a protein derived from Leptotrichia shahii, which recognizes a protospacer flanking sequence (PFS) (or RNA PAM (rPAM)) sequence motif of a single 3' A, U, or C, which may be located within the target nucleic acid.
  • PFS protospacer flanking sequence
  • rPAM RNA PAM
  • the CRISPR-Cas effector protein may be derived from Casl2a, which is a Type V Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-Cas nuclease see, e.g., SEQ ID NOs:l-20).
  • Casl2a differs in several respects from the more well- known Type II CRISPR Cas9 nuclease.
  • Cas9 recognizes a G-rich protospacer- adjacent motif (PAM) that is 3' to its guide RNA (gRNA, sgRNA, crRNA, crDNA, CRISPR array) binding site (protospacer, target nucleic acid, target DNA) (3'-NGG), while Casl2a recognizes a T-rich PAM that is located 5' to the target nucleic acid (5'-TTN, 5'-TTTN.
  • PAM G-rich protospacer- adjacent motif
  • Casl2a enzymes use a single guide RNA (gRNA, CRISPR array, crRNA) rather than the dual guide RNA (sgRNA (e.g., crRNA and tracrRNA)) found in natural Cas9 systems, and Casl2a processes its own gRNAs.
  • gRNA single guide RNA
  • crRNA dual guide RNA
  • Casl2a processes its own gRNAs.
  • Casl2a nuclease activity produces staggered DNA double stranded breaks instead of blunt ends produced by Cas9 nuclease activity, and Casl2a relies on a single RuvC domain to cleave both DNA strands, whereas Cas9 utilizes an HNH domain and a RuvC domain for cleavage.
  • a CRISPR Casl2a effector protein/domain useful with this invention may be any known or later identified Casl2a polypeptide (previously known as Cpfl) (see, e.g., U.S. Patent No. 9,790,490, which is incorporated by reference for its disclosures of Cpfl (Casl2a) sequences).
  • Casl2a refers to an RNA-guided nuclease comprising a Casl2a polypeptide, or a fragment thereof, which comprises the guide nucleic acid binding domain of Casl2a and/or an active, inactive, or partially active DNA cleavage domain of Casl2a.
  • a Casl2a useful with the invention may comprise a mutation in the nuclease active site (e.g., RuvC site of the Casl2a domain).
  • a Casl2a domain or Casl2a polypeptide having a mutation in its nuclease active site, and therefore, no longer comprising nuclease activity, is commonly referred to as deadCasl2a (e.g., dCasl2a).
  • deadCasl2a e.g., dCasl2a
  • a Casl2a domain or Casl2a polypeptide having a mutation in its nuclease active site may have impaired activity, e.g., may have nickase activity.
  • any deaminase domain/polypeptide useful for base editing may be used with this invention.
  • the deaminase domain may be a cytosine deaminase domain or an adenine deaminase domain.
  • a cytosine deaminase (or cytidine deaminase) useful with this invention may be any known or later identified cytosine deaminase from any organism (see, e.g., U.S. Patent No. 10,167,457 and Thuronyi et al. Nat. Biotechnol. 37:1070-1079 (2019), each of which is incorporated by reference herein for its disclosure of cytosine deaminases).
  • Cytosine deaminases can catalyze the hydrolytic deamination of cytidine or deoxy cytidine to uridine or deoxyuridine, respectively.
  • a deaminase or deaminase domain useful with this invention may be a cytidine deaminase domain, catalyzing the hydrolytic deamination of cytosine to uracil.
  • a cytosine deaminase may be a variant of a naturally-occurring cytosine deaminase, including but not limited to a primate (e.g., a human, monkey, chimpanzee, gorilla), a dog, a cow, a rat or a mouse.
  • a primate e.g., a human, monkey, chimpanzee, gorilla
  • a dog e.g., a cow, a rat or a mouse.
  • an cytosine deaminase useful with the invention may be about 70% to about 100% identical to a wild type cytosine deaminase (e.g., about 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical, and any range or value therein, to a naturally occurring cytosine deaminase).
  • a wild type cytosine deaminase e.g., about 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%,
  • a cytosine deaminase useful with the invention may be an apolipoprotein B mRNA-editing complex (APOBEC) family deaminase.
  • the cytosine deaminase may be an APOBEC 1 deaminase, an APOBEC2 deaminase, an APOBEC3A deaminase, an APOBEC3B deaminase, an APOBEC3C deaminase, an APOBEC3D deaminase, an APOBEC3F deaminase, an APOBEC3G deaminase, an APOBEC3H deaminase, an APOBEC4 deaminase, a human activation induced deaminase (hAID), an rAPOBECl, FERNY, and/or a CDA1, optionally a pmCDAl, an atC
  • APOBEC
  • the cytosine deaminase may be an APOBEC1 deaminase having the amino acid sequence of SEQ ID NO:23. In some embodiments, the cytosine deaminase may be an APOBEC3 A deaminase having the amino acid sequence of SEQ ID NO:24. In some embodiments, the cytosine deaminase may be an CDA1 deaminase, optionally a CDA1 having the amino acid sequence of SEQ ID NO:25. In some embodiments, the cytosine deaminase may be a FERNY deaminase, optionally a FERNY having the amino acid sequence of SEQ ID NO:26.
  • a cytosine deaminase useful with the invention may be about 70% to about 100% identical (e.g., 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or 100% identical) to the amino acid sequence of a naturally occurring cytosine deaminase (e.g., an evolved deaminase).
  • a naturally occurring cytosine deaminase e.g., an evolved deaminase
  • a cytosine deaminase useful with the invention may be about 70% to about 99.5% identical (e.g., about 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 99.5% identical) to the amino acid sequence of SEQ ID NO:23, SEQ ID NO:24, SEQ ID NO:25 or SEQ ID NO:26 (e.g., at least 80%, at least 85%, at least 90%, at least 92%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or at least 99.5% identical to the amino acid sequence of SEQ ID NO:23, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:26
  • a polynucleotide encoding a cytosine deaminase may be codon optimized for expression in a plant and the codon optimized polypeptide may be about 70% to 99.5% identical to the reference polynucleotide.
  • a nucleic acid construct of this invention may further encode a uracil glycosylase inhibitor (UGI) (e.g., uracil-DNA glycosylase inhibitor) polypeptide/domain.
  • UGI uracil glycosylase inhibitor
  • a nucleic acid construct encoding a CRISPR-Cas effector protein and a cytosine deaminase domain e.g., encoding a fusion protein comprising a CRISPR-Cas effector protein domain fused to a cytosine deaminase domain, and/or a CRISPR-Cas effector protein domain fused to a peptide tag or to an affinity polypeptide capable of binding a peptide tag and/or a deaminase protein domain fused to a peptide tag or to an affinity polypeptide capable of binding a peptide tag) may further encode a uracil-DNA glycosylase inhibitor (UGI), optionally wherein the
  • the invention provides fusion proteins comprising a CRISPR-Cas effector polypeptide, a deaminase domain, and a UGI and/or one or more polynucleotides encoding the same, optionally wherein the one or more polynucleotides may be codon optimized for expression in a plant.
  • the invention provides fusion proteins, wherein a CRISPR-Cas effector polypeptide, a deaminase domain, and a UGI may be fused to any combination of peptide tags and affinity polypeptides as described herein, thereby recruiting the deaminase domain and UGI to the CRISPR-Cas effector polypeptide and a target nucleic acid.
  • a guide nucleic acid may be linked to a recruiting RNA motif and one or more of the deaminase domain and/or UGI may be fused to an affinity polypeptide that is capable of interacting with the recruiting RNA motif, thereby recruiting the deaminase domain and UGI to a target nucleic acid.
  • a "uracil glycosylase inhibitor" useful with the invention may be any protein that is capable of inhibiting a uracil-DNA glycosylase base-excision repair enzyme.
  • a UGI domain comprises a wild type UGI or a fragment thereof.
  • a UGI domain useful with the invention may be about 70% to about 100% identical (e.g., 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or 100% identical and any range or value therein) to the amino acid sequence of a naturally occurring UGI domain.
  • a UGI domain may comprise the amino acid sequence of SEQ ID NO:41 or a polypeptide having about 70% to about 99.5% sequence identity to the amino acid sequence of SEQ ID NO:41 (e.g., at least 80%, at least 85%, at least 90%, at least 92%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or at least 99.5% identical to the amino acid sequence of SEQ ID NO:41).
  • a UGI domain may comprise a fragment of the amino acid sequence of SEQ ID NO:41 that is 100% identical to a portion of consecutive nucleotides (e.g., 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80 consecutive nucleotides; e.g., about 10, 15, 20, 25, 30, 35, 40, 45, to about 50, 55, 60, 65, 70, 75, 80 consecutive nucleotides) of the amino acid sequence of SEQ ID NO:41.
  • consecutive nucleotides e.g., 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80 consecutive nucleotides
  • a UGI domain may be a variant of a known UGI (e.g., SEQ ID NO:41) having about 70% to about 99.5% sequence identity (e.g., 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% sequence identity, and any range or value therein) to the known UGI.
  • sequence identity e.g., 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%
  • a polynucleotide encoding a UGI may be codon optimized for expression in a plant (e.g., a plant) and the codon optimized polypeptide may be about 70% to about 99.5% identical to the reference polynucleotide.
  • An adenine deaminase (or adenosine deaminase) useful with this invention may be any known or later identified adenine deaminase from any organism (see, e.g., U.S. Patent No.
  • An adenine deaminase can catalyze the hydrolytic deamination of adenine or adenosine.
  • the adenine deaminase may catalyze the hydrolytic deamination of adenosine or deoxy adenosine to inosine or deoxyinosine, respectively.
  • the adenosine deaminase may catalyze the hydrolytic deamination of adenine or adenosine in DNA.
  • an adenine deaminase encoded by a nucleic acid construct of the invention may generate an A G conversion in the sense (e.g., template) strand of the target nucleic acid or a T C conversion in the antisense (e.g., complementary) strand of the target nucleic acid.
  • an adenosine deaminase may be a variant of a naturally occurring adenine deaminase.
  • an adenosine deaminase may be about 70% to 100% identical to a wild type adenine deaminase (e.g., about 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%,
  • the deaminase or deaminase does not occur in nature and may be referred to as an engineered, mutated or evolved adenosine deaminase.
  • an engineered, mutated or evolved adenine deaminase polypeptide or an adenine deaminase domain may be about 70% to 99.9% identical to a naturally occurring adenine deaminase polypeptide/domain (e.g., about 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%,
  • the adenosine deaminase may be from a bacterium, (e.g., Escherichia coli, Staphylococcus aureus, Haemophilus influenzae, Caulobacter crescentus, and the like).
  • a polynucleotide encoding an adenine deaminase polypeptide/domain may be codon optimized for expression in a plant.
  • an adenine deaminase domain may be a wild type tRNA-specific adenosine deaminase domain, e.g., a tRNA-specific adenosine deaminase (TadA) and/or a mutated/evolved adenosine deaminase domain, e.g., mutated/evolved tRNA-specific adenosine deaminase domain (TadA*).
  • a TadA domain may be from E. coli.
  • the TadA may be modified, e.g., truncated, missing one or more N-terminal and/or C-terminal amino acids relative to a full-length TadA (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 6, 17, 18, 19, or 20 N-terminal and/or C terminal amino acid residues may be missing relative to a full length TadA.
  • a TadA polypeptide or TadA domain does not comprise an N-terminal methionine.
  • a wild type E. coli TadA comprises the amino acid sequence of SEQ ID NO:30.
  • a mutated/evolved comprises the amino acid sequence of SEQ ID NO:30.
  • coli TadA* comprises the amino acid sequence of SEQ ID NOs:31-40 (e.g., SEQ ID NOs:31, 32, 33, 34, 35, 36, 37, 38, 39 or 40).
  • a polynucleotide encoding a TadA/TadA* may be codon optimized for expression in a plant.
  • a cytosine deaminase catalyzes cytosine deamination and results in a thymidine (through a uracil intermediate), causing a C to T conversion, or a G to A conversion in the complementary strand in the genome.
  • the cytosine deaminase encoded by the polynucleotide of the invention generates a C T conversion in the sense (e.g., template) strand of the target nucleic acid or a G A conversion in antisense (e.g., complementary) strand of the target nucleic acid.
  • the adenine deaminase encoded by the nucleic acid construct of the invention generates an A G conversion in the sense (e.g., template) strand of the target nucleic acid or a T C conversion in the antisense (e.g., complementary) strand of the target nucleic acid.
  • nucleic acid constructs of the invention encoding a base editor comprising a sequence-specific DNA binding protein and a cytosine deaminase polypeptide, and nucleic acid constructs/expression cassettes/vectors encoding the same, may be used in combination with guide nucleic acids for modifying target nucleic acid including, but not limited to, generation of C T or G A mutations in a target nucleic acid including, but not limited to, a plasmid sequence; generation of C T or G A mutations in a coding sequence to alter an amino acid identity; generation of C T or G A mutations in a coding sequence to generate a stop codon; generation of C T or G A mutations in a coding sequence to disrupt a start codon; generation of point mutations in genomic DNA to disrupt function; and/or generation of point mutations in genomic DNA to disrupt splice junctions.
  • nucleic acid constructs of the invention encoding a base editor comprising a sequence-specific DNA binding protein and an adenine deaminase polypeptide, and expression cassettes and/or vectors encoding the same may be used in combination with guide nucleic acids for modifying a target nucleic acid including, but not limited to, generation of A G or T C mutations in a target nucleic acid including, but not limited to, a plasmid sequence; generation of A G or T C mutations in a coding sequence to alter an amino acid identity; generation of A G or T C mutations in a coding sequence to generate a stop codon; generation of A G or T C mutations in a coding sequence to disrupt a start codon; generation of point mutations in genomic DNA to disrupt function; and/or generation of point mutations in genomic DNA to disrupt splice junctions.
  • the nucleic acid constructs of the invention comprising a CRISPR-Cas effector protein or a fusion protein thereof may be used in combination with a guide RNA (gRNA, CRISPR array, CRISPR RNA, crRNA), designed to function with the encoded CRISPR-Cas effector protein or domain, to modify a target nucleic acid.
  • a guide RNA gRNA, CRISPR array, CRISPR RNA, crRNA
  • a guide nucleic acid useful with this invention comprises at least one spacer sequence and at least one repeat sequence.
  • the guide nucleic acid is capable of forming a complex with the CRISPR-Cas nuclease domain encoded and expressed by a nucleic acid construct of the invention and the spacer sequence is capable of hybridizing to a target nucleic acid, thereby guiding the complex (e.g., a CRISPR-Cas effector fusion protein (e.g., CRISPR-Cas effector domain fused to a deaminase domain and/or a CRISPR-Cas effector domain fused to a peptide tag or an affinity polypeptide to recruit a deaminase domain and optionally, a UGI) to the target nucleic acid, wherein the target nucleic acid may be modified (e.g., cleaved or edited) or modulated (e.g., modulating transcription) by the deaminase domain.
  • a CRISPR-Cas effector fusion protein e.g., CRISPR-Cas effector
  • a nucleic acid construct encoding a Cas9 domain linked to a cytosine deaminase domain may be used in combination with a Cas9 guide nucleic acid to modify a target nucleic acid, wherein the cytosine deaminase domain of the fusion protein deaminates a cytosine base in the target nucleic acid, thereby editing the target nucleic acid.
  • a nucleic acid construct encoding a Cas9 domain linked to an adenine deaminase domain may be used in combination with a Cas9 guide nucleic acid to modify a target nucleic acid, wherein the adenine deaminase domain of the fusion protein deaminates an adenosine base in the target nucleic acid, thereby editing the target nucleic acid.
  • a nucleic acid construct encoding a Casl2a domain (or other selected CRISPR-Cas nuclease, e.g., C2cl, C2c3, Casl2b, Casl2c, Casl2d, Casl2e, Casl3a, Casl3b, Casl3c, Casl3d, Casl, CaslB, Cas2, Cas3, Cas3', Cas3", Cas4, Cas5, Cas6, Cas7, Cas8, Cas9 (also known as Csnl and Csxl2), CaslO, Csyl, Csy2, Csy3, Csel, Cse2, Cscl, Csc2, Csa5, Csn2, Csm2, Csm3, Csm4, Csm5, Csm6, Cmrl, Cmr3, Cmr4, Cmr5, Cmr6, Csbl, Csb2, Cs
  • Csxl4, CsxlO, Csxl6, CsaX, Csx3, Csxl, Csxl5, Csfl, Csf2, Csf3, Csf4 (dinG), and/or Csf5) linked to a cytosine deaminase domain or adenine deaminase domain may be used in combination with a Casl2a guide nucleic acid (or the guide nucleic acid for the other selected CRISPR-Cas nuclease) to modify a target nucleic acid, wherein the cytosine deaminase domain or adenine deaminase domain of the fusion protein deaminates a cytosine base in the target nucleic acid, thereby editing the target nucleic acid.
  • a Casl2a guide nucleic acid or the guide nucleic acid for the other selected CRISPR-Cas nuclease
  • a “guide nucleic acid,” “guide RNA,” “gRNA,” “CRISPR RNA/DNA” “crRNA” or “crDNA” as used herein means a nucleic acid that comprises at least one spacer sequence, which is complementary to (and hybridizes to) a target DNA (e.g., protospacer), and at least one repeat sequence (e.g., a repeat of a Type V Casl2a CRISPR-Cas system, or a fragment or portion thereof; a repeat of a Type II Cas9 CRISPR-Cas system, or fragment thereof; a repeat of a Type V C2cl CRISPR Cas system, or a fragment thereof; a repeat of a CRISPR-Cas system of, for example, C2c3, Casl2a (also referred to as Cpfl), Casl2b, Casl2c, Casl2d, Casl2e, Casl3a, Casl3b, Casl3c, Cas
  • a Cas 12a gRNA may comprise, from 5' to 3', a repeat sequence (full length or portion thereof ("handle”); e.g., pseudoknot-like structure) and a spacer sequence.
  • a guide nucleic acid may comprise more than one repeat sequence-spacer sequence (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, or more repeat-spacer sequences) (e.g., repeat-spacer-repeat, e.g., repeat-spacer-repeat-spacer-repeat-spacer-repeat-spacer-repeat-spacer-repeat-spacer, and the like).
  • the guide nucleic acids of this invention are synthetic, human-made and not found in nature.
  • a gRNA can be quite long and may be used as an aptamer (like in the MS2 recruitment strategy) or other RNA structures hanging off the spacer.
  • a "repeat sequence” as used herein refers to, for example, any repeat sequence of a wild-type CRISPR Cas locus (e.g., a Cas9 locus, a Casl2a locus, a C2cl locus, etc.) or a repeat sequence of a synthetic crRNA that is functional with the CRISPR-Cas effector protein encoded by the nucleic acid constructs of the invention.
  • a repeat sequence useful with this invention can be any known or later identified repeat sequence of a CRISPR-Cas locus (e.g., Type I, Type II, Type III, Type IV, Type V or Type VI) or it can be a synthetic repeat designed to function in a
  • a repeat sequence may comprise a hairpin structure and/or a stem loop structure.
  • a repeat sequence may form a pseudoknot-bke structure at its 5' end (i.e., "handle").
  • a repeat sequence can be identical to or substantially identical to a repeat sequence from wild-type Type I CRISPR-Cas loci, Type II, CRISPR-Cas loci, Type III, CRISPR-Cas loci, Type IV CRISPR-Cas loci, Type V CRISPR-Cas loci and/or Type VI CRISPR-Cas loci.
  • a repeat sequence from a wild-type CRISPR-Cas locus may be determined through established algorithms, such as using the CRISPRfinder offered through CRISPRdb (see, Grissa et al. Nucleic Acids Res. 35(Web Server issue):W52-7).
  • a repeat sequence or portion thereof is linked at its 3' end to the 5' end of a spacer sequence, thereby forming a repeat-spacer sequence (e.g., guide nucleic acid, guide RNA/DNA, crRNA, crDNA).
  • a repeat sequence comprises, consists essentially of, or consists of at least 10 nucleotides depending on the particular repeat and whether the guide nucleic acid comprising the repeat is processed or unprocessed (e.g., about 10, 11, 12, 13, 14, 15, 16, 17, 18,
  • a repeat sequence comprises, consists essentially of, or consists of about 10 to about 20, about 10 to about 30, about 10 to about 45, about 10 to about 50, about 15 to about 30, about 15 to about 40, about 15 to about 45, about 15 to about 50, about 20 to about 30, about 20 to about 40, about 20 to about 50, about 30 to about 40, about 40 to about 80, about 50 to about 100 or more nucleotides.
  • a repeat sequence linked to the 5' end of a spacer sequence can comprise a portion of a repeat sequence (e.g., 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 or more contiguous nucleotides of a wild type repeat sequence).
  • a portion of a repeat sequence linked to the 5' end of a spacer sequence can be about five to about ten consecutive nucleotides in length (e.g., about 5, 6, 7, 8, 9, 10 nucleotides) and have at least 90% sequence identity (e.g., at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more (e.g., 99.1, 99.2, 99.3, 99.4, 99.5, 99.6, 99.7, 99.8, 99.9, or 100%)) to the same region (e.g., 5' end) of a wild type CRISPR Cas repeat nucleotide sequence.
  • a portion of a repeat sequence may comprise a pseudoknot-bke structure at its 5' end (e.g., "handle").
  • a "spacer sequence” as used herein is a nucleotide sequence that is complementary to a target nucleic acid (e.g., target DNA) (e.g., protospacer) (e.g., consecutive nucleotides of a sequence (a) encoding an amino acid sequence having at least 70% sequence identity to SEQ ID NO: 1
  • a spacer sequence may include, but is not limited to, the nucleotide sequences of any one of SEQ ID NOs:126-130.
  • the spacer sequence can be fully complementary or substantially complementary (e.g., at least about 70% complementary (e.g., about 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more (e.g., 99.1, 99.2, 99.3, 99.4, 99.5, 99.6, 99.7, 99.8, 99.9, or 100%)) to a target nucleic acid.
  • 70% complementary e.g., about 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%,
  • the spacer sequence can have one, two, three, four, or five mismatches as compared to the target nucleic acid, which mismatches can be contiguous or noncontiguous.
  • the spacer sequence can have 70% complementarity to a target nucleic acid.
  • the spacer nucleotide sequence can have 80% complementarity to a target nucleic acid.
  • the spacer nucleotide sequence can have 85%, 90%, 95%, 96%, 97%, 98%, 99% or 99.5% complementarity, and the like, to the target nucleic acid (protospacer).
  • the spacer sequence is 100% complementary to the target nucleic acid.
  • a spacer sequence may have a length from about 15 nucleotides to about 30 nucleotides (e.g., 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleotides, or any range or value therein).
  • a spacer sequence may have complete complementarity or substantial complementarity over a region of a target nucleic acid (e.g., protospacer) that is at least about 15 nucleotides to about 30 nucleotides in length.
  • the spacer is about 20 nucleotides in length.
  • the spacer is about 21, 22, or 23 nucleotides in length.
  • the 5' region of a spacer sequence of a guide nucleic acid may be identical to a target DNA, while the 3' region of the spacer may be substantially complementary to the target DNA (e.g., Type V CRISPR-Cas), or the 3' region of a spacer sequence of a guide nucleic acid may be identical to a target DNA, while the 5' region of the spacer may be substantially complementary to the target DNA (e.g., Type II CRISPR-Cas), and therefore, the overall complementarity of the spacer sequence to the target DNA may be less than 100%.
  • the target DNA e.g., Type V CRISPR-Cas
  • the first 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 nucleotides in the 5' region (i.e., seed region) of, for example, a 20 nucleotide spacer sequence may be 100% complementary to the target DNA, while the remaining nucleotides in the 3' region of the spacer sequence are substantially complementary (e.g., at least about 70% complementary) to the target DNA.
  • the first 1 to 8 nucleotides (e.g., the first 1, 2, 3, 4, 5, 6, 7, 8, nucleotides, and any range therein) of the 5' end of the spacer sequence may be 100% complementary to the target DNA, while the remaining nucleotides in the 3' region of the spacer sequence are substantially complementary (e.g., at least about 50% complementary (e.g., 50%, 55%, 60%, 65%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%,
  • the first 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 nucleotides in the 3' region (i.e., seed region) of, for example, a 20 nucleotide spacer sequence may be 100% complementary to the target DNA, while the remaining nucleotides in the 5' region of the spacer sequence are substantially complementary (e.g., at least about 70% complementary) to the target DNA.
  • the first 1 to 10 nucleotides (e.g., the first 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 nucleotides, and any range therein) of the 3' end of the spacer sequence may be 100% complementary to the target DNA, while the remaining nucleotides in the 5' region of the spacer sequence are substantially complementary (e.g., at least about 50% complementary (e.g., at least about 50%, 55%, 60%, 65%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more or any range or value therein)) to the target DNA.
  • the remaining nucleotides in the 5' region of the spacer sequence are substantially complementary (e.g., at least about 50% complementary (e.g., at
  • a seed region of a spacer may be about 8 to about 10 nucleotides in length, about 5 to about 6 nucleotides in length, or about 6 nucleotides in length.
  • a “target nucleic acid”, “target DNA,” “target nucleotide sequence,” “target region,” or a “target region in the genome” refers to a region of a plant's genome that is fully complementary (100% complementary) or substantially complementary (e.g., at least 70% complementary (e.g., 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%,
  • a target region useful for a CRISPR-Cas system may be located immediately 3' (e.g., Type V CRISPR- Cas system) or immediately 5' (e.g., Type II CRISPR-Cas system) to a PAM sequence in the genome of the organism (e.g., a plant genome).
  • a target region may be selected from any region of at least 15 consecutive nucleotides (e.g., 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30 nucleotides, and the like) located immediately adjacent to a PAM sequence.
  • a "protospacer sequence” refers to the target double stranded DNA and specifically to the portion of the target DNA (e.g., or target region in the genome) that is fully or substantially complementary (and hybridizes) to the spacer sequence of the CRISPR repeat-spacer sequences (e.g., guide nucleic acids, CRISPR arrays, crRNAs).
  • Type V CRISPR-Cas e.g., Casl2a
  • Type II CRISPR-Cas Cas9
  • the protospacer sequence is flanked by (e.g., immediately adjacent to) a protospacer adjacent motif (PAM).
  • PAM protospacer adjacent motif
  • Type IV CRISPR-Cas systems the PAM is located at the 5' end on the non-target strand and at the 3' end of the target strand (see below, as an example).
  • Type II CRISPR-Cas e.g., Cas9
  • the PAM is located immediately 3' of the target region.
  • the PAM for Type I CRISPR-Cas systems is located 5' of the target strand.
  • Canonical Casl2a PAMs are T rich.
  • a canonical Casl2a PAM sequence may be 5'-TTN, 5'-TTTN, or 5'-TTTV.
  • canonical Cas9 (e.g., S. pyogenes) PAMs may be 5'-NGG ⁇ 3'.
  • non-canonical PAMs may be used but may be less efficient.
  • Additional PAM sequences may be determined by those skilled in the art through established experimental and computational approaches.
  • experimental approaches include targeting a sequence flanked by all possible nucleotide sequences and identifying sequence members that do not undergo targeting, such as through the transformation of target plasmid DNA (Esvelt et al. 2013. Nat. Methods 10:1116-1121; Jiang et al. 2013. Nat. Biotechnol. 31:233-239).
  • a computational approach can include performing BLAST searches of natural spacers to identify the original target DNA sequences in bacteriophages or plasmids and aligning these sequences to determine conserved sequences adjacent to the target sequence (Briner and Barrangou. 2014. Appl. Environ. Microbiol. 80:994- 1001; Mojica et al. 2009. Microbiology 155:733-740).
  • the present invention provides expression cassettes and/or vectors comprising the nucleic acid constructs of the invention (e.g., one or more components of an editing system of the invention).
  • expression cassettes and/or vectors comprising the nucleic acid constructs of the invention and/or one or more guide nucleic acids may be provided.
  • a nucleic acid construct of the invention encoding a base editor e.g., a construct comprising a CRISPR-Cas effector protein and a deaminase domain (e.g., a fusion protein)
  • the components for base editing e.g., a CRISPR-Cas effector protein fused to a peptide tag or an affinity polypeptide, a deaminase domain fused to a peptide tag or an affinity polypeptide, and/or a UGI fused to a peptide tag or an affinity polypeptide
  • a base editor e.g., a construct comprising a CRISPR-Cas effector protein and a deaminase domain (e.g., a fusion protein)
  • the components for base editing e.g., a CRISPR-Cas effector protein fused to a peptide tag or an affinity polypeptide, a deaminase domain fused to
  • a target nucleic acid may be contacted with (e.g., provided with) the expression cassette(s) or vector(s) encoding the base editor or components for base editing in any order from one another and the guide nucleic acid, e.g., prior to, concurrently with, or after the expression cassette comprising the guide nucleic acid is provided (e.g., contacted with the target nucleic acid).
  • Fusion proteins of the invention may comprise sequence-specific nucleic acid binding domains, CRISPR-Cas polypeptides, and/or deaminase domains fused to peptide tags or affinity polypeptides that interact with the peptide tags, as known in the art, for use in recruiting the deaminase to the target nucleic acid.
  • Methods of recruiting may also comprise guide nucleic acids linked to RNA recruiting motifs and deaminases fused to affinity polypeptides capable of interacting with RNA recruiting motifs, thereby recruiting the deaminase to the target nucleic acid.
  • chemical interactions may be used to recruit polypeptides (e.g., deaminases) to a target nucleic acid.
  • a peptide tag (e.g., epitope) useful with this invention may include, but is not limited to, a GCN4 peptide tag (e.g., Sun-Tag), a c-Myc affinity tag, an HA affinity tag, a His affinity tag, an S affinity tag, a methionine-His affinity tag, an RGD-His affinity tag, a FLAG octapeptide, a strep tag or strep tag II, a V5 tag, and/or a VS V -G epitope.
  • GCN4 peptide tag e.g., Sun-Tag
  • a c-Myc affinity tag e.g., an HA affinity tag, a His affinity tag, an S affinity tag, a methionine-His affinity tag, an RGD-His affinity tag, a FLAG octapeptide, a strep tag or strep tag II, a V5 tag, and/or a
  • a peptide tag may comprise 1 or 2 or more copies of a peptide tag (e.g., repeat unit, multimerized epitope (e.g., tandem repeats)) (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25 or more repeat units.
  • an affinity polypeptide that interacts with/binds to a peptide tag may be an antibody.
  • the antibody may be a scFv antibody.
  • an affinity polypeptide that binds to a peptide tag may be synthetic (e.g., evolved for affinity interaction) including, but not limited to, an affibody, an anticalin, a monobody and/or a DARPin (see, e.g., Sha et ak, Protein Sci. 26(5):910-924 (2017)); Gilbreth ( Curr Opin Struc Biol 22(4):413-420 (2013)), U.S. Patent No. 9,982,053, each of which are incorporated by reference in their entireties for the teachings relevant to affibodies, anticalins, monobodies and/or DARPins.
  • Example peptide tag sequences and their affinity polypeptides include, but are not limited to, the amino acid sequences of SEQ
  • a guide nucleic acid may be linked to an RNA recruiting motif, and a polypeptide to be recruited (e.g., a deaminase) may be fused to an affinity polypeptide that binds to the RNA recruiting motif, wherein the guide binds to the target nucleic acid and the RNA recruiting motif binds to the affinity polypeptide, thereby recruiting the polypeptide to the guide and contacting the target nucleic acid with the polypeptide (e.g., deaminase).
  • two or more polypeptides may be recruited to a guide nucleic acid, thereby contacting the target nucleic acid with two or more polypeptides (e.g., deaminases).
  • Example RNA recruiting motifs and their affinity polypeptides include, but are not limited to, the sequences of SEQ ID NOs:48-58.
  • a polypeptide fused to an affinity polypeptide may be a reverse transcriptase and the guide nucleic acid may be an extended guide nucleic acid linked to an RNA recruiting motif.
  • an RNA recruiting motif may be located on the 3' end of the extended portion of an extended guide nucleic acid (e.g., 5'-3', repeat-spacer- extended portion (RT template-primer binding site)-RNA recruiting motii).
  • an RNA recruiting motif may be embedded in the extended portion.
  • an extended guide RNA and/or guide RNA may be linked to one or to two or more RNA recruiting motifs (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more motifs, e.g., at least 10 to about 25 motifs, e.g., about 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25 motifs), optionally wherein when two or more RNA recruiting motifs are present they may be the same RNA recruiting motif or different RNA recruiting motifs.
  • RNA recruiting motifs e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more motifs, e.g., at least 10 to about 25 motifs, e.g., about 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25 motifs
  • an RNA recruiting motif and corresponding affinity polypeptide may include, but is not limited, to a telomerase Ku binding motif (e.g., Ku binding hairpin) and the corresponding affinity polypeptide Ku (e.g., Ku heterodimer), a telomerase Sm7 binding motif and the corresponding affinity polypeptide Sm7, an MS2 phage operator stem-loop and the corresponding affinity polypeptide MS2 Coat Protein (MCP), a PP7 phage operator stem-loop and the corresponding affinity polypeptide PP7 Coat Protein (PCP), an SfMu phage Com stem- loop and the corresponding affinity polypeptide Com RNA binding protein, a PUF binding site (PBS) and the affinity polypeptide Pumilio/fem-3 mRNA binding factor (PUF), and/or a synthetic RNA-aptamer and the aptamer ligand as the corresponding affinity polypeptide.
  • a telomerase Ku binding motif e.g., Ku binding hairpin
  • the RNA recruiting motif and corresponding affinity polypeptide may be an MS2 phage operator stem-loop and the affinity polypeptide MS2 Coat Protein (MCP).
  • MCP MS2 Coat Protein
  • the RNA recruiting motif and corresponding affinity polypeptide may be a PUF binding site (PBS) and the affinity polypeptide Pumilio/fem-3 mRNA binding factor (PUF).
  • the components for recruiting polypeptides and nucleic acids may those that function through chemical interactions that may include, but are not limited to, rapamycin-inducible dimerization of FRB - FKBP; Biotin-streptavidin; SNAP tag; Halo tag; CLIP tag; DmrA-DmrC heterodimer induced by a compound; bifunctional ligand (e.g., fusion of two protein-binding chemicals together, e.g., dihyrofolate reductase (DHFR).
  • rapamycin-inducible dimerization of FRB - FKBP Biotin-streptavidin
  • SNAP tag Halo tag
  • CLIP tag DmrA-DmrC heterodimer induced by a compound
  • bifunctional ligand e.g., fusion of two protein-binding chemicals together, e.g., dihyrofolate reductase (DHFR).
  • the nucleic acid constructs, expression cassettes or vectors of the invention that are optimized for expression in a plant may be about 70% to 100% identical (e.g., about 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or 100%) to the nucleic acid constructs, expression cassettes or vectors comprising the same polynucleotide(s) but which have not been codon optimized for expression in a plant.
  • cells comprising one or more polynucleotides, guide nucleic acids, nucleic acid constructs, expression cassettes or vectors of the invention.
  • nucleic acid constructs of the invention e.g., a construct comprising a sequence specific nucleic acid binding domain, a CRISPR-Cas effector domain, a deaminase domain, reverse transcriptase (RT), RT template and/or a guide nucleic acid, etc.
  • expression cassettes/vectors comprising the same may be used as an editing system of this invention for modifying target nucleic acids and/or their expression.
  • plants or plant cultivars which are to be treated with preference in accordance with the invention include all plants which, through genetic modification, received genetic material, which imparts particular advantageous useful properties ("traits") to these plants.
  • advantageous useful properties are better plant growth, vigor, stress tolerance, standability, lodging resistance, nutrient uptake, plant nutrition, and/or yield, in particular improved growth, increased tolerance to high or low temperatures, increased tolerance to drought or to levels of water or soil salinity, enhanced flowering performance, easier harvesting, accelerated ripening, higher yields, higher quality and/or a higher nutritional value of the harvested products, better storage life and/or processability of the harvested products.
  • Such properties are an increased resistance against animal and microbial pests, such as against insects, arachnids, nematodes, mites, slugs and snails owing, for example, to toxins formed in the plants.
  • animal and microbial pests such as against insects, arachnids, nematodes, mites, slugs and snails owing, for example, to toxins formed in the plants.
  • DNA sequences encoding proteins which confer properties of tolerance to such animal and microbial pests, in particular insects mention will particularly be made of the genetic material from Bacillus thuringiensis encoding the Bt proteins widely described in the literature and well known to those skilled in the art. Mention will also be made of proteins extracted from bacteria such as Photorhabdus (W097/17432 and WO98/08932).
  • Bt Cry or VIP proteins which include the CrylA, CrylAb, CrylAc, CryllA, CrylllA, CryIIIB2, Cry9c Cry2Ab, Cry3Bb and CrylF proteins or toxic fragments thereof and also hybrids or combinations thereof, especially the CrylF protein or hybrids derived from a CrylF protein (e.g. hybrid CrylA-CrylF proteins or toxic fragments thereof), the CrylA-type proteins or toxic fragments thereof, preferably the CrylAc protein or hybrids derived from the CrylAc protein (e.g.
  • hybrid CrylAb-CrylAc proteins or the CrylAb or Bt2 protein or toxic fragments thereof, the Cry2Ae, Cry2Af or Cry2Ag proteins or toxic fragments thereof, the CrylA.105 protein or a toxic fragment thereof, the VIP3Aal9 protein, the VIP3Aa20 protein, the VIP3 A proteins produced in the COT202 or COT203 cotton events, the VIP3 Aa protein ora toxic fragment thereof as described in Estruch et al. (1996), Proc Natl Acad Sci US A.
  • Another and particularly emphasized example of such properties is conferred tolerance to one or more herbicides, for example imidazolinones, sulphonylureas, glyphosate or phosphinothricin.
  • herbicides for example imidazolinones, sulphonylureas, glyphosate or phosphinothricin.
  • DNA sequences encoding proteins i.e., polynucleotides of interest
  • the bar or PAT gene or the Streptomyces coelicolor gene described in W02009/152359 which confers tolerance to glufosinate herbicides
  • a gene encoding a suitable EPSPS (5-Enolpyruvylshikimat-3-phosphat-Synthase) which confers tolerance to herbicides having EPSPS as a target, especially herbicides such as glyphosate and its salts, a gene encoding glyphosate-n-acetyltrans
  • Patent 6,855,533 genes encoding 2,4-D-monooxygenases conferring tolerance to 2,4-D (2,4- dichlorophenoxyacetic acid) and genes encoding Dicamba monooxygenases conferring tolerance to dicamba (3,6-dichloro-2- methoxybenzoic acid).
  • Such properties are increased resistance against phytopathogenic fungi, bacteria and/or viruses owing, for example, to systemic acquired resistance (SAR), systemin, phytoalexins, elicitors and also resistance genes and correspondingly expressed proteins and toxins.
  • SAR systemic acquired resistance
  • systemin phytoalexins
  • elicitors resistance genes and correspondingly expressed proteins and toxins.
  • Particularly useful transgenic events in transgenic plants or plant cultivars which can be treated with preference in accordance with the invention include Event 531/ PV-GHBK04 (cotton, insect control, described in W02002/040677), Event 1143-14A (cotton, insect control, not deposited, described in WO2006/128569); Event 1143-51B (cotton, insect control, not deposited, described in W02006/128570); Event 1445 (cotton, herbicide tolerance, not deposited, described in US-A 2002-120964 or W02002/034946); Event 17053 (rice, herbicide tolerance, deposited as PTA-9843, described in WO2010/117737); Event 17314 (rice, herbicide tolerance, deposited as PTA-9844, described in WO2010/117735); Event 281-24-236 (cotton, insect control - herbicide tolerance, deposited as PTA-6233, described in W02005/103266 or US-A 2005-216969); Event 3006-210-23 (cotton, insect control - herbicide
  • Event BLR1 (oilseed rape, restoration of male sterility, deposited as NCIMB 41193, described in W02005/074671), Event CE43-67B (cotton, insect control, deposited as DSM ACC2724, described in US-A 2009-217423 or WO2006/128573); Event CE44-69D (cotton, insect control, not deposited, described in US-A 2010- 0024077); Event CE44-69D (cotton, insect control, not deposited, described in WO2006/128571); Event CE46-02A (cotton, insect control, not deposited, described in WO2006/128572); Event COT102 (cotton, insect control, not deposited, described in US-A 2006-130175 or W02004/039986);
  • Event COT202 (cotton, insect control, not deposited, described in US-A 2007-067868 or W02005/054479); Event COT203 (cotton, insect control, not deposited, described in W02005/054480); ); Event DAS21606-3 / 1606 (soybean, herbicide tolerance, deposited as PTA-11028, described in WO2012/033794), Event DAS40278 (com, herbicide tolerance, deposited as ATCC PTA-10244, described in WO2011/022469); Event DAS-44406-6 / pDAB8264.44.06.1 (soybean, herbicide tolerance, deposited as PTA-11336, described in WO2012/075426), Event DAS-14536-7 /pDAB8291.45.36.2 (soybean, herbicide tolerance, deposited as PTA-11335, described in WO2012/075429), Event DAS-59122-7 (com, insect control - herbicide tolerance, deposited as ATCC PTA
  • Event NK603 (com, herbicide tolerance, deposited as ATCC PTA-2478, described in US-A 2007-292854); Event PE-7 (rice, insect control, not deposited, described in W02008/114282); Event RF3 (oilseed rape, pollination control - herbicide tolerance, deposited as ATCC PTA-730, described in W02001/041558 or US-A 2003- 188347); Event RT73 (oilseed rape, herbicide tolerance, not deposited, described in W02002/036831 or US-A 2008-070260); Event SYHT0H2 / SYN-000H2-5 (soybean, herbicide tolerance, deposited as PTA-11226, described in WO2012/082548), Event T227-1 (sugar beet, herbicide tolerance, not deposited, described in W02002/44407 or US-A 2009-265817); Event T25 (com, herbicide tolerance, not
  • the genes/events may also be present in combinations with one another in the transgenic plants.
  • transgenic plants which may be mentioned are the important crop plants, such as cereals (wheat, rice, triticale, barley, rye, oats), maize, soya beans, potatoes, sugar beet, sugar cane, tomatoes, peas and other types of vegetable, cotton, tobacco, oilseed rape and also fruit plants (with the fruits apples, pears, citrus fruits and grapes), with particular emphasis being given to maize, soya beans, wheat, rice, potatoes, cotton, sugar cane, tobacco and oilseed rape.
  • Traits which are particularly emphasized are the increased resistance of the plants to insects, arachnids, nematodes and slugs and snails, as well as the increased resistance of the plants to one or more herbicides.
  • CRN genes are used to create alleles of the CRN gene that reduce signaling and increase kernel row number (KRN) without compromising ear organization (e.g., without substantially reducing ear length).
  • the CRN gene encodes an inactive pseudo-kinase with a short extracellular (EC) domain. See Fig. 1.
  • the EC of CRN is required for plasma membrane transiting but is not required for dimerization with CLV2, suggesting that the EC domain may perform a charge masking function.
  • CRN genes are modified to produce CRN polypeptide mutations that may provide a reduction in positive charge, and thus, less charge masking of FEA2 and greater complex retention in the ER.
  • the genomic sequence of the CRN gene was identified in a proprietary maize line. From this reference sequence, spacer sequences (SEQ ID NOs: 126-130) were designed for use in editing constructs.
  • the editing constructs contained a CRISPR-Cas effector and a spacer sequence designed to target the EC domain of the CRN protein encoded by the CRN gene. Spacers were deployed with either a Cas-effector cutting enzyme or a Cas-effector base editing complex.
  • a vector encoding a selected spacer as well as the chosen CRISPR-Cas effector were introduced into dried excised maize embryos using Agrobacterium. Transformed tissue was maintained in vitro with antibiotic selection to regenerate positive transformants. Healthy non chimeric plants (E0) were selected and plugged in growth trays. Tissue was collected from regenerating plants (E0 generation) for DNA extraction and subsequent molecular screening was employed to identify edits in the CRN gene.
  • Fig. 2 shows some of the edits obtained and their position within the EC domain of the CRN gene. Plants identified to be (1) healthy, non chimeric and fertile, with (2) low transgene copy and (3) a deletion in the EC domain were advanced to the next generation. E0 plants that satisfied all the above criteria were selfed to produce the El generation. Selected Els were selfed to generate the E2 generation. We identified several families with deletions in and around the EC domain.
  • Seeds for El and E2 material were sown in flats and later transferred to pots after seedlings were established. All materials were cultivated under standard greenhouse conditions and grown to reproductive maturity. Following standard practices, emerging ears were covered with small paper bags prior to the emergence of silk and tassels were covered during anthesis for the capture of pollen on a plant-by-plant basis. All ears were removed from the plants after dry- down. After ear harvest and dry-down, kernel row number was manually counted for all ears. Data represent the average of three row counts per ear taken from the mid-section of the ear where row lineages were most defined.
  • a marker e.g., paper clip
  • Un-edited germplasm, and lines transformed with a Gus plasmid were used as wild-type controls for phenotyping.
  • Table 1 shows kernel row number (KRN) for El and E2 families derived from selfed E0 and selfed El ears, respectively.
  • BCF1 ears are a cross of edited El plants with a WT inbred different than the original WT transformation line.
  • the edit is provided with reference to the CRN gene represented by SEQ ID
  • the full-length sequence for each edited gene can be determined by locating within SEQ ID NO: 122 the named base pair position under column labeled "allele" and delete the number of base pairs (nucleotides) provided (e.g., 964:9D is a 9 base pair deletion starting at position 964 of SEQ ID NO: 122).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Organic Chemistry (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Molecular Biology (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • Plant Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Biophysics (AREA)
  • Cell Biology (AREA)
  • Botany (AREA)
  • Developmental Biology & Embryology (AREA)
  • Environmental Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Natural Medicines & Medicinal Plants (AREA)
  • Physiology (AREA)
  • Breeding Of Plants And Reproduction By Means Of Culturing (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Image Processing (AREA)
  • Paper (AREA)

Abstract

La présente invention concerne des compositions et des procédés de modification de gènes CORYNE (CRN) chez des végétaux pour augmenter éventuellement le nombre de rangées de grains et/ou améliorer un ou plusieurs aspects du rendement. L'invention concerne en outre des végétaux ayant un nombre de rangées de grains augmenté et/ou un ou plusieurs aspects de rendement améliorés, produits à l'aide des procédés et des compositions de l'invention.
EP21735063.6A 2020-06-02 2021-06-01 Procédés de régulation de la taille du méristème pour l'amélioration des cultures Pending EP4156909A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202063033273P 2020-06-02 2020-06-02
PCT/US2021/035114 WO2021247477A1 (fr) 2020-06-02 2021-06-01 Procédés de régulation de la taille du méristème pour l'amélioration des cultures

Publications (1)

Publication Number Publication Date
EP4156909A1 true EP4156909A1 (fr) 2023-04-05

Family

ID=76624202

Family Applications (1)

Application Number Title Priority Date Filing Date
EP21735063.6A Pending EP4156909A1 (fr) 2020-06-02 2021-06-01 Procédés de régulation de la taille du méristème pour l'amélioration des cultures

Country Status (9)

Country Link
US (1) US20210371873A1 (fr)
EP (1) EP4156909A1 (fr)
CN (1) CN116096230A (fr)
AR (1) AR122533A1 (fr)
BR (1) BR112022024489A2 (fr)
CA (1) CA3185017A1 (fr)
MX (1) MX2022015107A (fr)
UY (1) UY39250A (fr)
WO (1) WO2021247477A1 (fr)

Family Cites Families (120)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2003A (en) 1841-03-12 Improvement in horizontal windivhlls
US97687A (en) 1869-12-07 Improvement in feed-cutter
US2010A (en) 1841-03-18 Machine foe
US24077A (en) 1859-05-17 Window-sash supporter
US2009A (en) 1841-03-18 Improvement in machines for boring war-rockets
US137395A (en) 1873-04-01 Improvement in nuts
NZ221259A (en) 1986-07-31 1990-05-28 Calgene Inc Seed specific transcriptional regulation
US6040504A (en) 1987-11-18 2000-03-21 Novartis Finance Corporation Cotton promoter
CA1339684C (fr) 1988-05-17 1998-02-24 Peter H. Quail Systeme de collecte de l'ubiquitine vegetale
US5641876A (en) 1990-01-05 1997-06-24 Cornell Research Foundation, Inc. Rice actin gene and promoter
EP0452269B1 (fr) 1990-04-12 2002-10-09 Syngenta Participations AG Promoteurs à préférence tissulaire
US6395966B1 (en) 1990-08-09 2002-05-28 Dekalb Genetics Corp. Fertile transgenic maize plants containing a gene encoding the pat protein
US5459252A (en) 1991-01-31 1995-10-17 North Carolina State University Root specific gene promoter
DE69230290T2 (de) 1991-08-27 2000-07-20 Novartis Ag Proteine mit insektiziden eigenschaften gegen homopteran insekten und ihre verwendung im pflanzenschutz
UA48104C2 (uk) 1991-10-04 2002-08-15 Новартіс Аг Фрагмент днк, який містить послідовність,що кодує інсектицидний протеїн, оптимізовану для кукурудзи,фрагмент днк, який забезпечує направлену бажану для серцевини стебла експресію зв'язаного з нею структурного гена в рослині, фрагмент днк, який забезпечує специфічну для пилку експресію зв`язаного з нею структурного гена в рослині, рекомбінантна молекула днк, спосіб одержання оптимізованої для кукурудзи кодуючої послідовності інсектицидного протеїну, спосіб захисту рослин кукурудзи щонайменше від однієї комахи-шкідника
PL186091B1 (pl) 1995-04-20 2003-10-31 American Cyanamid Co Wyizolowany DNA, wektor, komórka, warianty białkaAHAS, sposób nadawania oporności na herbicydy komórce, sposób wytwarzania opornego na herbicydy białka oraz sposoby zwalczania chwastów
MX9705101A (es) 1995-11-06 1997-10-31 Wisconsin Alumni Res Found Toxinas proteinicas insecticidas obtenidas a partir de photorhabdus luminescens.
CA2263819A1 (fr) 1996-08-29 1998-03-05 Dow Agrosciences Llc Toxines proteiniques insecticides isolees a partir de photorhabdus
EP0975778B8 (fr) 1997-04-03 2007-11-21 DeKalb Genetics Corporation Utilisation de lignees de mais resistantes aux glyphosates
IL128165A0 (en) 1997-05-05 1999-11-30 Dow Agrosciences Llc Insecticidal protein toxins from xenorhabdus
CN1301300A (zh) 1998-02-20 2001-06-27 曾尼卡有限公司 花粉特异性启动子
AU1336200A (en) 1998-11-03 2000-05-22 Aventis Cropscience N.V. Glufosinate tolerant rice
US6333449B1 (en) 1998-11-03 2001-12-25 Plant Genetic Systems, N.V. Glufosinate tolerant rice
US6509516B1 (en) 1999-10-29 2003-01-21 Plant Genetic Systems N.V. Male-sterile brassica plants and methods for producing same
US6506963B1 (en) 1999-12-08 2003-01-14 Plant Genetic Systems, N.V. Hybrid winter oilseed rape and methods for producing same
ATE421973T1 (de) 1999-12-28 2009-02-15 Bayer Bioscience Nv Inzektizide proteinen von bacillus thuringiensis
US6395485B1 (en) 2000-01-11 2002-05-28 Aventis Cropscience N.V. Methods and kits for identifying elite event GAT-ZM1 in biological samples
MXPA02009335A (es) 2000-03-27 2003-02-12 Syngenta Participations Ag Promotores del virus de la rizadura amarilla del cestrum.
AR044724A1 (es) 2000-03-27 2005-10-05 Syngenta Participations Ag Promotores del virus de la rizadura amarilla del cestrum
BRPI0100752B1 (pt) 2000-06-22 2015-10-13 Monsanto Co moléculas e pares de moléculas de dna, processos para detectar molécula de dna e para criar um traço tolerante a glifosato em plantas de milho, bem como kit de detecção de dna
US6713259B2 (en) 2000-09-13 2004-03-30 Monsanto Technology Llc Corn event MON810 and compositions and methods for detection thereof
WO2002027004A2 (fr) 2000-09-29 2002-04-04 Monsanto Technology Llc Plante de ble 33391 resistante au glyphosate et compositions et procedes de detection de celle-ci
WO2002034946A2 (fr) 2000-10-25 2002-05-02 Monsanto Technology Llc Mecanisme biochimique de plant de coton pv-ghgt07(1445), compositions et techniques de detection de celui-ci
AU3089902A (en) 2000-10-30 2002-05-15 Monsanto Technology Llc Canola event pv-bngt04(rt73) and compositions and methods for detection thereof
AR035215A1 (es) 2000-11-20 2004-05-05 Monsanto Technology Llc Polinucleotido aislado, primer y segundo polinucleotido cebador, metodo para detectar el suceso vegetal de algodon 531, molecula de polinucleotido aislado obtenida por dicho metodo, equipo de deteccion de acido nucleico y metodo para determinar la cigosidad del genoma de una planta de algodon.
EP1337669A2 (fr) 2000-11-30 2003-08-27 Ses Europe N.V./S.A. Betterave sucriere transgenique resistante au glyphosate characterisee par une insertion specifique du transgene (t227-1), procede et amorces pour la detection de cette insertion.
EG26529A (en) 2001-06-11 2014-01-27 مونسانتو تكنولوجى ل ل سى Prefixes for detection of DNA molecule in cotton plant MON15985 which gives resistance to damage caused by insect of squamous lepidoptera
US6818807B2 (en) 2001-08-06 2004-11-16 Bayer Bioscience N.V. Herbicide tolerant cotton plants having event EE-GH1
AR037856A1 (es) 2001-12-17 2004-12-09 Syngenta Participations Ag Evento de maiz
WO2004011601A2 (fr) 2002-07-29 2004-02-05 Monsanto Technology, Llc Mais pv-zmir13 designe mon863, composition et procedes de detection
GB0225129D0 (en) 2002-10-29 2002-12-11 Syngenta Participations Ag Improvements in or relating to organic compounds
CN100469885C (zh) 2002-12-05 2009-03-18 孟山都技术有限公司 翦股颖事件asr-368和组合物及其检测方法
AU2004211592B2 (en) 2003-02-12 2008-04-10 Monsanto Technology Llc Cotton event MON 88913 and compositions and methods for detection thereof
US7335816B2 (en) 2003-02-28 2008-02-26 Kws Saat Ag Glyphosate tolerant sugar beet
SI1597373T1 (sl) 2003-02-20 2012-11-30 Kws Saat Ag Sladkorna pesa odporna proti glifosatu
JP2006525028A (ja) 2003-05-02 2006-11-09 ダウ・アグロサイエンシーズ・エルエルシー トウモロコシイベントtc1507およびそれを検出するための方法
US7129397B2 (en) 2003-10-06 2006-10-31 Syngenta Participations Ag Promoters functional in plant plastids
KR100537955B1 (ko) 2003-10-29 2005-12-20 학교법인고려중앙학원 꽃가루 특이적 유전자 발현 프로모터
AU2004295386A1 (en) 2003-12-01 2005-06-16 Syngenta Participations Ag Insect resistant cotton plants and methods of detecting the same
WO2005054480A2 (fr) 2003-12-01 2005-06-16 Syngenta Participations Ag Plants de coton resistant aux insectes et procedes de detection de ces derniers
US7157281B2 (en) 2003-12-11 2007-01-02 Monsanto Technology Llc High lysine maize compositions and event LY038 maize plants
PT1708560E (pt) 2003-12-15 2015-06-03 Monsanto Technology Llc Planta de milho mon88017 e composições e métodos para a sua detecção
GB0402106D0 (en) 2004-01-30 2004-03-03 Syngenta Participations Ag Improved fertility restoration for ogura cytoplasmic male sterile brassica and method
PT1737290E (pt) 2004-03-25 2015-07-02 Syngenta Participations Ag Evento do milho mir604
BRPI0418683B8 (pt) 2004-03-26 2022-06-28 Dow Agrosciences Llc Polinucleotìdeos referentes aos eventos cry1f 281-24-236 e cry1ac 3006-210-23
AR050891A1 (es) 2004-09-29 2006-11-29 Du Pont Evento das-59122-7 de maiz y metodos para su deteccion
EP1868426B1 (fr) 2005-03-16 2018-02-21 Syngenta Participations AG Mais 3272 et procedes pour le detecter
ES2388548T3 (es) 2005-04-08 2012-10-16 Bayer Cropscience Nv Suceso de élite A2704-12 y métodos y estuches para identificar a dicho suceso en muestras biológicas
CN105112520B (zh) 2005-04-11 2019-05-28 拜尔作物科学公司 原种事件a5547-127以及在生物样品中鉴定此类事件的方法和试剂盒
AP2693A (en) 2005-05-27 2013-07-16 Monsanto Technology Llc Soybean event MON89788 and methods for detection thereof
WO2006128572A1 (fr) 2005-06-02 2006-12-07 Syngenta Participations Ag Coton insecticide ce46-02a
WO2006128570A1 (fr) 2005-06-02 2006-12-07 Syngenta Participations Ag Coton insecticide 1143-51b
US20100024077A1 (en) 2005-06-02 2010-01-28 Syngenta Participations Ag Ce44-69d insecticidal cotton
WO2006128569A2 (fr) 2005-06-02 2006-12-07 Syngenta Participations Ag Coton insecticide 1143-14a
WO2006128568A2 (fr) 2005-06-02 2006-12-07 Syngenta Participations Ag Coton insecticide t342-142
AU2006254493B2 (en) 2005-06-02 2010-12-09 Syngenta Participations Ag CE43- 67B, insecticidal transgenic cotton expressing CRY1AB
US7932439B2 (en) 2005-08-08 2011-04-26 Bayer Bioscience N.V. Herbicide tolerant cotton plants and methods for identifying the same
AR055128A1 (es) 2005-08-24 2007-08-08 Du Pont Metodos y composiciones para la expresion de un polinucleotido de interes
JP5164862B2 (ja) 2006-02-10 2013-03-21 マハラシュートラ ハイブリッド シーズ カンパニー リミテッド(マヒコ) Ee−1イベントを含むトランスジェニックナス(solanummelongena)
WO2007140256A1 (fr) 2006-05-26 2007-12-06 Monsanto Technology, Llc Plant et semence de maïs correspondant au produit transgénique mon89034, procédés de détection et utilisation associés
EA021187B1 (ru) 2006-06-03 2015-04-30 Зингента Партисипейшнс Аг Хромосомный целевой сайт кукурузы и способ получения трансгенной кукурузы
US7951995B2 (en) 2006-06-28 2011-05-31 Pioneer Hi-Bred International, Inc. Soybean event 3560.4.3.5 and compositions and methods for the identification and detection thereof
US7928295B2 (en) 2006-08-24 2011-04-19 Bayer Bioscience N.V. Herbicide tolerant rice plants and methods for identifying same
US20080064032A1 (en) 2006-09-13 2008-03-13 Syngenta Participations Ag Polynucleotides and uses thereof
US7928296B2 (en) 2006-10-30 2011-04-19 Pioneer Hi-Bred International, Inc. Maize event DP-098140-6 and compositions and methods for the identification and/or detection thereof
EP2078089B1 (fr) 2006-10-31 2016-05-04 E. I. du Pont de Nemours and Company Événement de soja dp-305423-1, leurs compositions et leurs procédés d'identification et/ou de détection
WO2008114282A2 (fr) 2007-03-19 2008-09-25 Maharashtra Hybrid Seeds Company Limited Riz transgénique (oryza sativa) comprenant l'événement pe-7 et son procédé de détection
ES2432406T3 (es) 2007-04-05 2013-12-03 Bayer Cropscience Nv Plantas de algodón resistentes a los insectos y métodos para identificación de las mismas
US8309818B2 (en) 2007-06-11 2012-11-13 Bayer Cropscience B.V. Insect resistant cotton plants and methods for identifying same
BR122017018105B1 (pt) 2007-11-15 2024-01-23 Monsanto Technology Llc Molécula de dna genômico de soja transgênica
WO2009100188A2 (fr) 2008-02-08 2009-08-13 Dow Agrosciences Llc Procédés de détection de l’événement de maïs das-59132
CA2715564C (fr) 2008-02-14 2018-01-09 E. I. Du Pont De Nemours And Company Evenement spt flanquant l'adn genomique vegetal et procedes d'identification de l'evenement spt
JP5767813B2 (ja) 2008-02-15 2015-08-19 モンサント テクノロジー エルエルシー トランスジェニック事象mon87769に対応するダイズ植物および種子ならびにそれらを検出する方法
AR070495A1 (es) 2008-02-29 2010-04-07 Monsanto Technology Llc Evento de maiz mon87460 y composiciones y metodos para detectarlo
AU2009257375B2 (en) 2008-06-11 2016-07-07 Dow Agrosciences Llc Constructs for expressing herbicide tolerance genes, related plants, and related trait combinations
US9078406B2 (en) 2008-08-29 2015-07-14 Monsanto Technology Llc Soybean plant and seed corresponding to transgenic event MON87754 and methods for detection thereof
MX2011003297A (es) 2008-09-29 2011-04-21 Monsanto Technology Llc Evento transgenico de frijol de soya mon87705 y metodos para deteccion del mismo.
MX346319B (es) 2008-12-16 2017-03-15 Syngenta Participations Ag Evento 5307 del maiz.
EA201100945A1 (ru) 2008-12-19 2012-01-30 Зингента Партисипейшнс Аг Трансгенный вариант сахарной свеклы gm rz13
CA2748973A1 (fr) 2009-01-07 2010-07-15 Basf Agrochemical Products B.V. Evenement de soja 127 et procedes apparentes
WO2010117737A1 (fr) 2009-03-30 2010-10-14 Monsanto Technology Llc Evénement de riz transgénique 17053 et ses procédés d'utilisation
WO2010117735A1 (fr) 2009-03-30 2010-10-14 Monsanto Technology Llc Évènement 17314 de riz transgénique et ses procédés d'utilisation
NZ598599A (en) 2009-08-19 2014-06-27 Dow Agrosciences Llc Aad-1 event das-40278-9, related transgenic corn lines, and event-specific identification thereof
EP3875592A3 (fr) 2009-09-17 2021-11-03 Monsanto Technology LLC Variété transgénique mon 87708 du soja et ses méthodes d'utilisation
PL2503873T3 (pl) 2009-11-23 2017-09-29 Bayer Cropscience Nv Rośliny soi tolerujące herbicyd i sposoby ich identyfikowania
WO2011062904A1 (fr) 2009-11-23 2011-05-26 Monsanto Technology Llc Événement du maïs transgénique mon 87427 et échelle de développement relative
US8581046B2 (en) 2010-11-24 2013-11-12 Pioneer Hi-Bred International, Inc. Brassica gat event DP-073496-4 and compositions and methods for the identification and/or detection thereof
CN104928256A (zh) 2009-11-24 2015-09-23 陶氏益农公司 Aad-12事件416、相关的转基因大豆系及其事件特异性鉴定
BR112012012494A2 (pt) 2009-11-24 2020-11-03 Dow Agrosciences Llc detecção de evento de soja aad-12 416
WO2011075595A1 (fr) 2009-12-17 2011-06-23 Pioneer Hi-Bred International, Inc. Maïs dp-043a47-3 et procédés de détection associés
PL2512226T3 (pl) 2009-12-17 2019-10-31 Pioneer Hi Bred Int Modyfikacja DP-004114-3 kukurydzy i sposoby jej wykrywania
US20110154525A1 (en) 2009-12-17 2011-06-23 Pioneer Hi-Bred International, Inc. Maize event DP-040416-8 and methods for detection thereof
WO2011084632A1 (fr) 2009-12-17 2011-07-14 Pioneer Hi-Bred International, Inc. Evénement de maïs dp-032316-8 et ses procédés de détection
AU2011261580B2 (en) 2010-06-04 2013-12-19 Monsanto Technology Llc Transgenic brassica event MON 88302 and methods of use thereof
US8785728B2 (en) 2010-09-08 2014-07-22 Dow Agrosciences, Llc. AAD-12 event 1606 and related transgenic soybean lines
EP2627785A4 (fr) 2010-10-12 2014-03-19 Monsanto Technology Llc Plante et semence de soja correspondant à l'événement transgénique mon87712 et procédé pour les détecter
WO2012071039A1 (fr) 2010-11-24 2012-05-31 Pioner Hi-Bred International, Inc. Événement dp-061061-7 de brassica gat et compositions et procédés pour l'identifier et/ou le détecter
CA2819684C (fr) 2010-12-03 2024-05-07 Dow Agrosciences Llc Evenement 8264.44.06.1 de tolerance aux herbicides empile, lignees de soja transgeniques apparentees, et sa detection
BR112013015745B1 (pt) 2010-12-03 2021-01-19 Ms Technologies, Llc polinucleotídeos referente ao evento de tolerância a herbicida 8291.45.36.2, cassete de expressão, sonda, bem como processos para identificação do evento, determinação da zigosidade, produção de uma planta de soja transgênica e produção de uma proteína em uma célula de planta
TWI667347B (zh) 2010-12-15 2019-08-01 瑞士商先正達合夥公司 大豆品種syht0h2及偵測其之組合物及方法
ES2743728T3 (es) 2011-03-30 2020-02-20 Monsanto Technology Llc Evento transgénico MON 88701 del algodón y procedimientos de uso del mismo
EP3228713A1 (fr) 2011-06-30 2017-10-11 Monsanto Technology LLC Plante et graine de luzerne correspondant à l'événement transgénique kk 179-2 et procédés pour la détection de celui-ci
RU2636021C2 (ru) 2011-07-13 2017-11-17 ДАУ АГРОСАЙЕНСИЗ ЭлЭлСи Линии трансгенной сои, генетическое событие 8264.42.32.1, устойчивое к гербицидам с пакетированными генами на его основе, и их детектирование
WO2013012643A1 (fr) 2011-07-15 2013-01-24 Syngenta Participations Ag Polynucléotides codant pour la tréhalose-6-phosphate phosphatase et procédés d'utilisation de ceux-ci
AP2014007885A0 (en) 2012-02-01 2014-08-31 Dow Agrosciences Llc Novel class of glyphosate resistance genes
AU2015298356B2 (en) 2014-08-05 2020-11-19 MabQuest SA Immunological reagents binding to pd-1
US9790490B2 (en) 2015-06-18 2017-10-17 The Broad Institute Inc. CRISPR enzymes and systems
JP7109784B2 (ja) 2015-10-23 2022-08-01 プレジデント アンド フェローズ オブ ハーバード カレッジ 遺伝子編集のための進化したCas9蛋白質
EP3494215A1 (fr) 2016-08-03 2019-06-12 President and Fellows of Harvard College Éditeurs de nucléobases d'adénosine et utilisations associées
CA3105847A1 (fr) * 2018-07-23 2020-01-30 Pioneer Hi-Bred International, Inc. Procedes et compositions pour augmenter le rendement au moyen de modifications du locus genomique fea3 et ligands associes

Also Published As

Publication number Publication date
CN116096230A (zh) 2023-05-09
MX2022015107A (es) 2023-03-01
UY39250A (es) 2021-12-31
AR122533A1 (es) 2022-09-21
WO2021247477A1 (fr) 2021-12-09
BR112022024489A2 (pt) 2023-01-31
CA3185017A1 (fr) 2021-12-09
US20210371873A1 (en) 2021-12-02

Similar Documents

Publication Publication Date Title
US20220411813A1 (en) Modification of growth regulating factor family transcription factors in soybean
AU2022352997A1 (en) Methods and compositions for reducing pod shatter in canola
US20230212600A1 (en) Methods for controlling meristem size for crop improvement
WO2023019188A1 (fr) Modification de gènes du récepteur des brassinostéroïdes pour améliorer des caractéristiques de rendement
EP4291641A1 (fr) Procédés et compositions pour modifier les niveaux de cytokinine oxydase dans des plantes
US20210371873A1 (en) Methods for controlling meristem size for crop improvement
US20210395767A1 (en) Methods for controlling meristem size for crop improvement
WO2024006791A1 (fr) Procédés et compositions permettant de réguler la taille du méristème pour améliorer les cultures
WO2024006792A1 (fr) Procédés et compositions permettant de réguler la taille du méristème pour améliorer les cultures
WO2023192838A1 (fr) Plantes rosacées à floraison précoce présentant des caractéristiques améliorées
WO2023196886A1 (fr) Procédés et compositions pour améliorer la résistance à la fusariose de l'épi
WO2024036240A1 (fr) Procédés et compositions pour réguler la taille des méristèmes pour l'amélioration des cultures
WO2024030984A1 (fr) Procédés et compositions pour améliorer des caractères de rendement
WO2024054880A1 (fr) Procédés et compositions pour améliorer des caractéristiques de rendement dans des végétaux
WO2022182834A1 (fr) Procédés et compositions pour modifier l'architecture racinaire chez des plantes
WO2023034731A1 (fr) Modification de gènes de peptidase de liaison à l'ubiquitine dans des plantes pour améliorer des caractéristiques de rendement
WO2023168217A1 (fr) Modification de gènes récepteurs de brassinostéroïdes pour améliorer des caractéristiques de rendement
US20230348922A1 (en) Methods and compositions for enhancing yield and disease resistance
WO2023034891A1 (fr) Procédés et compositions pour améliorer l'architecture des plantes et les caractéristiques de rendement
WO2023060028A1 (fr) Procédés d'amélioration de la fertilité du fleuron et du rendement en graines
WO2023023496A1 (fr) Procédés et compositions pour modifier les gènes de l'histidine kinase du récepteur de la cytokinine dans des plantes
WO2023205714A1 (fr) Procédés et compositions pour améliorer des caractéristiques de rendement

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: UNKNOWN

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20221220

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)